1
|
Trouvé P, Férec C. p.Phe508del-CFTR Trafficking: A Protein Quality Control Perspective Through UPR, UPS, and Autophagy. Int J Mol Sci 2025; 26:3623. [PMID: 40332143 PMCID: PMC12026709 DOI: 10.3390/ijms26083623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Cystic fibrosis (CF) is a genetic disease due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most frequent mutation (p.Phe508del) results in a misfolded protein (p.Phe508del-CFTR) with an altered transport to the membrane of the cells via the conventional protein secretion (CPS) pathway. Nevertheless, it can use unconventional protein secretion (UPS). Indeed, p.Phe508del-CFTR forms a complex with GRASP55 to assist its direct trafficking from the endoplasmic reticulum to the plasma membrane. While GRASP55 is a key player of UPS, it is also a key player of stress-induced autophagy. In parallel, the unfolded protein response (UPR), which is activated in the presence of misfolded proteins, is tightly linked to UPS and autophagy through the key effectors IRE1, PERK, and ATF6. A better understanding of how UPS, UPR, and stress-induced autophagy interact to manage protein trafficking in CF and other conditions could lead to novel therapeutic strategies. By enhancing or modulating these pathways, it may be possible to increase p.Phe508del-CFTR surface expression. In summary, this review highlights the critical roles of UPS- and UPR-induced autophagy in managing protein transport, offering new perspectives for therapeutic approaches.
Collapse
Affiliation(s)
- Pascal Trouvé
- Univ Brest, Inserm, EFS, UMR 1078, 22 Avenue Camille Desmoulins, F-29200 Brest, France;
| | | |
Collapse
|
2
|
Saar D, Lennartsson CLE, Weidner P, Burgermeister E, Kragelund BB. The Myotubularin Related Proteins and the Untapped Interaction Potential of Their Disordered C-Terminal Regions. Proteins 2025; 93:831-854. [PMID: 39614773 PMCID: PMC11878207 DOI: 10.1002/prot.26774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 03/05/2025]
Abstract
Intrinsically disordered regions (IDRs) of proteins remain understudied with enigmatic sequence features relevant to their functions. Members of the myotubularin-related protein (MTMR) family contain uncharacterized IDRs. After decades of research on their phosphatase activity, recent work on the C-terminal IDRs of MTMR7 revealed new interactions and important new functions beyond the phosphatase function. Here we take a broader look at the C-terminal domains (CTDs) of 14 human MTMRs and use bioinformatic tools and biophysical methods to ask which other functions may be probable in this protein family. The predictions show that the CTDs are disordered and carry short linear motifs (SLiMs) important for targeting of MTMRs to defined subcellular compartments and implicating them in signaling, phase separation, interaction with diverse proteins, including transcription factors and are of relevance for cancer research and neuroscience. We also present experimental methods to study the CTDs and use them to characterize the coiled coil (CC) domains of MTMR7 and MTMR9. We show homo- and hetero-oligomerization with preference for MTMR7-CC to form dimers, while MTMR9-CC forms trimers. We relate the results to sequence features and make predictions for the structural landscape of other MTMRs. Our work gives a broad insight into the so far unrecognized features and SLiMs in MTMR-CTDs, and provides the basis for more in-depth experimental research on this diverse protein family and understudied IDRs in proteins in general.
Collapse
Affiliation(s)
- Daniel Saar
- REPINUniversity of CopenhagenCopenhagenDenmark
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | | | - Philip Weidner
- Department of Medicine II, University Medical Center MannheimHeidelberg UniversityMannheimGermany
| | - Elke Burgermeister
- Department of Medicine II, University Medical Center MannheimHeidelberg UniversityMannheimGermany
| | - Birthe B. Kragelund
- REPINUniversity of CopenhagenCopenhagenDenmark
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
3
|
Mella J, Volk R, Zaro B, Buchwalter A. C-terminal tagging, transmembrane domain hydrophobicity, and an ER retention motif influence the secretory trafficking of the inner nuclear membrane protein emerin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633811. [PMID: 39896633 PMCID: PMC11785080 DOI: 10.1101/2025.01.19.633811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The inner nuclear membrane (INM), a subdomain of the endoplasmic reticulum (ER), sequesters hundreds of transmembrane proteins within the nucleus. We previously found that one INM protein, emerin, can evade the INM by secretory transport to the lysosome, where it is degraded. In this work, we used targeted mutagenesis to identify intrinsic sequences that promote or inhibit emerin's secretory trafficking. By manipulating these sequences across several tag and expression level combinations, we now find that emerin's localization is sensitive to C-terminal GFP tagging. While emerin's long, hydrophobic C-terminal transmembrane domain facilitates trafficking to the lysosome, extending its lumenal terminus with a GFP tag biases the protein toward this pathway. In contrast, we identify a conserved ER retention sequence that stabilizes N- and C-terminally tagged emerin by limiting its lysosomal flux. These findings underscore long-standing concerns about tagging artifacts and reveal novel determinants of tail-anchored INM protein targeting.
Collapse
Affiliation(s)
- Jessica Mella
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Regan Volk
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Balyn Zaro
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
4
|
Foo B, Amedei H, Kaur S, Jaawan S, Boshnakovska A, Gall T, de Boer RA, Silljé HHW, Urlaub H, Rehling P, Lenz C, Lehnart SE. Unbiased complexome profiling and global proteomics analysis reveals mitochondrial impairment and potential changes at the intercalated disk in presymptomatic R14Δ/+ mice hearts. PLoS One 2024; 19:e0311203. [PMID: 39446877 PMCID: PMC11501035 DOI: 10.1371/journal.pone.0311203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/15/2024] [Indexed: 10/26/2024] Open
Abstract
Phospholamban (PLN) is a sarco-endoplasmic reticulum (SER) membrane protein that regulates cardiac contraction/relaxation by reversibly inhibiting the SERCA2a Ca2+-reuptake pump. The R14Δ-PLN mutation causes severe cardiomyopathy that is resistant to conventional treatment. Protein complexes and higher-order supercomplexes such as intercalated disk components and Ca+2-cycling domains underlie many critical cardiac functions, a subset of which may be disrupted by R14Δ-PLN. Complexome profiling (CP) is a proteomics workflow for systematic analysis of high molecular weight (MW) protein complexes and supercomplexes. We hypothesize that R14Δ-PLN may alter a subset of these assemblies, and apply CP workflows to explore these changes in presymptomatic R14Δ/+ mice hearts. Ventricular tissues from presymptomatic 28wk-old WT and R14Δ/+ mice were homogenized under non-denaturing conditions, fractionated by size-exclusion chromatography (SEC) with a linear MW-range exceeding 5 MDa, and subjected to quantitative data-independent acquisition mass spectrometry (DIA-MS) analysis. Unfortunately, current workflows for the systematic analysis of CP data proved ill-suited for use in cardiac samples. Most rely upon curated protein complex databases to provide ground-truth for analysis; however, these are derived primarily from cancerous or immortalized cell lines and, consequently, cell-type specific complexes (including cardiac-specific machinery potentially affected in R14Δ-PLN hearts) are poorly covered. We thus developed PERCOM: a novel CP data-analysis strategy that does not rely upon these databases and can, furthermore, be implemented on widely available spreadsheet software. Applying PERCOM to our CP dataset resulted in the identification of 296 proteins with disrupted elution profiles. Hits were significantly enriched for mitochondrial and intercalated disk (ICD) supercomplex components. Changes to mitochondrial supercomplexes were associated with reduced expression of mitochondrial proteins and maximal oxygen consumption rate. The observed alterations to mitochondrial and ICD supercomplexes were replicated in a second cohort of "juvenile" 9wk-old mice. These early-stage changes to key cardiac machinery may contribute to R14Δ-PLN pathogenesis.
Collapse
Affiliation(s)
- Brian Foo
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Hugo Amedei
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Surmeet Kaur
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Samir Jaawan
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Tanja Gall
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, Rotterdam, the Netherlands
| | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henning Urlaub
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Christof Lenz
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stephan E. Lehnart
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Matczuk AK, Kublicka A, Chodaczek G, Siedlecka M. Dual topology of equine arteritis virus GP3 protein and the role of arginine motif RXR in GP3 ER retention. Virology 2024; 597:110122. [PMID: 38850896 DOI: 10.1016/j.virol.2024.110122] [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: 12/18/2023] [Revised: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024]
Abstract
Glycoprotein 3 (GP3) serves as a structural protein in equine arteritis virus (EAV), forming a heterotrimeric complex that plays a pivotal role in virus tropism. In this study, we tested the membrane topology of GP3, both when expressed separately and during infection with recombinant tagged EAV GP3-HA. In our antibody accessibility experiment, we made a noteworthy discovery: GP3, when expressed separately, exhibits a dual topology. We introduced an additional N-glycosylation site, which was only partially used, providing further evidence for the dual topology of GP3. Intriguingly, this mutated GP3 was secreted into the medium, a result of the disruption of the ER retention motif RXR. The additional glycosylation site was not used when we examined the recombinant EAV virus with the same mutation. Despite the fact of higher expression levels of mutant GP3-HA, the protein was not secreted, and the recombinant mutant virus did not have growth delay compared to the EAV wild-type virus. This finding suggests that GP3 has a single type one membrane topology in virus infected cells, whereas the expression of GP3 in trans results in the dual topology of this protein. The RXR motif in the C-terminus is a co-factor of ER retention of the protein, but the main retention signal remains elusive.
Collapse
Affiliation(s)
- Anna K Matczuk
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland.
| | - Agata Kublicka
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Grzegorz Chodaczek
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Magdalena Siedlecka
- Department of Epizootiology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| |
Collapse
|
6
|
Ali H, Noyvert D, Hankinson J, Lindsey G, Lulla A, Lulla V. The astrovirus N-terminal nonstructural protein anchors replication complexes to the perinuclear ER membranes. PLoS Pathog 2024; 20:e1011959. [PMID: 39008516 PMCID: PMC11271882 DOI: 10.1371/journal.ppat.1011959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/25/2024] [Accepted: 06/20/2024] [Indexed: 07/17/2024] Open
Abstract
An essential aspect of positive-sense RNA virus replication is anchoring the replication complex (RC) to cellular membranes. Positive-sense RNA viruses employ diverse strategies, including co-translational membrane targeting through signal peptides and co-opting cellular membrane trafficking components. Often, N-terminal nonstructural proteins play a crucial role in linking the RC to membranes, facilitating the early association of the replication machinery. Astroviruses utilize a polyprotein strategy to synthesize nonstructural proteins, relying on subsequent processing to form replication-competent complexes. This study provides evidence for the perinuclear ER membrane association of RCs in five distinct human astrovirus strains. Using tagged recombinant classical human astrovirus 1 and neurotropic MLB2 strains, we establish that the N-terminal domain guides the ER membrane association. We identified di-arginine motifs responsible for the perinuclear ER retention and formation of functional RCs through mutational analysis of the N-terminal domain in replicon and reverse genetics systems. In addition, we demonstrate the association of key components of the astrovirus replication complex: double-stranded RNA, RNA-dependent RNA polymerase, protease, and N-terminal protein. Our findings highlight the intricate virus-ER interaction mechanism employed by astroviruses, potentially leading to the development of novel antiviral intervention strategies.
Collapse
Affiliation(s)
- Hashim Ali
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - David Noyvert
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Gemma Lindsey
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Aleksei Lulla
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Valeria Lulla
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
7
|
Sharma PK, Weng JH, Manschwetus JT, Wu J, Ma W, Herberg FW, Taylor SS. Role of the leucine-rich repeat protein kinase 2 C-terminal tail in domain cross-talk. Biochem J 2024; 481:313-327. [PMID: 38305364 PMCID: PMC10903466 DOI: 10.1042/bcj20230477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/03/2024]
Abstract
Leucine-rich repeat protein kinase 2 (LRRK2) is a multi-domain protein encompassing two of biology's most critical molecular switches, a kinase and a GTPase, and mutations in LRRK2 are key players in the pathogenesis of Parkinson's disease (PD). The availability of multiple structures (full-length and truncated) has opened doors to explore intra-domain cross-talk in LRRK2. A helix extending from the WD40 domain and stably docking onto the kinase domain is common in all available structures. This C-terminal (Ct) helix is a hub of phosphorylation and organelle-localization motifs and thus serves as a multi-functional protein : protein interaction module. To examine its intra-domain interactions, we have recombinantly expressed a stable Ct motif (residues 2480-2527) and used peptide arrays to identify specific binding sites. We have identified a potential interaction site between the Ct helix and a loop in the CORB domain (CORB loop) using a combination of Gaussian accelerated molecular dynamics simulations and peptide arrays. This Ct-Motif contains two auto-phosphorylation sites (T2483 and T2524), and T2524 is a 14-3-3 binding site. The Ct helix, CORB loop, and the CORB-kinase linker together form a part of a dynamic 'CAP' that regulates the N-lobe of the kinase domain. We hypothesize that in inactive, full-length LRRK2, the Ct-helix will also mediate interactions with the N-terminal armadillo, ankyrin, and LRR domains (NTDs) and that binding of Rab substrates, PD mutations, or kinase inhibitors will unleash the NTDs.
Collapse
Affiliation(s)
- Pallavi Kaila Sharma
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093-0652, U.S.A
| | - Jui-Hung Weng
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093-0652, U.S.A
| | - Jascha T. Manschwetus
- Department of Biochemistry, University of Kassel, Heinrich-Plett-Str. 40, 34132 Kassel, Hessen, Germany
| | - Jian Wu
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093-0652, U.S.A
| | - Wen Ma
- Department of Physics, University of Vermont, Burlington, Vermont
| | - Friedrich W. Herberg
- Department of Biochemistry, University of Kassel, Heinrich-Plett-Str. 40, 34132 Kassel, Hessen, Germany
| | - Susan S. Taylor
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093-0652, U.S.A
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0652, U.S.A
| |
Collapse
|
8
|
Wu Y, Chen X, Zhu L, Wang D, Li X, Song J, Wang D, Yu X, Li Y, Tang BZ. Endoplasmic Reticulum-Targeted Aggregation-Induced Emission Luminogen for Synergetic Tumor Ablation with Glibenclamide. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37903083 DOI: 10.1021/acsami.3c10940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Photodynamic therapy based on fluorescence illumination of subcellular organelles and in situ bursts of reactive oxygen species (ROS) has been recognized as a promising strategy for cancer theranostics. However, the short life of ROS and unclarified anticancer mechanism seriously restrict the application. Herein, we rationally designed and facilely synthesized a 2,6-dimethylpyridine-based triphenylamine (TPA) derivative TPA-DMPy with aggregation-induced emission (AIE) features and production of type-I ROS. Except for its selective binding to the endoplasmic reticulum (ER), TPA-DMPy, in synergy with glibenclamide, a medicinal agent used against diabetes, induced significant apoptosis of cancer cells in vitro and in vivo. Additionally, TPA-DMPy greatly incited the release of calcium from ER upon light irradiation to further aggravate the depolarization of ER membrane potential caused by glibenclamide, thus inducing fatal ER stress and crosstalk between ER and mitochondria. Our study extends the biological design and application of AIE luminogens and provides new insights into discovering novel anticancer targets and agents.
Collapse
Affiliation(s)
- Yifan Wu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xiaohui Chen
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Liwei Zhu
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Deliang Wang
- Department of Materials Chemistry, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Xue Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jiayi Song
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xiyong Yu
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Ying Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Ben Zhong Tang
- Shenzhen Institute of Aggregate Science and Engineering, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| |
Collapse
|
9
|
Zhu X, Yin J, Guo H, Wang Y, Ma B. Vesicle trafficking in rice: too little is known. FRONTIERS IN PLANT SCIENCE 2023; 14:1263966. [PMID: 37790794 PMCID: PMC10543891 DOI: 10.3389/fpls.2023.1263966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023]
Abstract
The vesicle trafficking apparatus is a fundamental machinery to maintain the homeostasis of membrane-enclosed organelles in eukaryotic cells. Thus, it is broadly conserved in eukaryotes including plants. Intensive studies in the model organisms have produced a comprehensive picture of vesicle trafficking in yeast and human. However, with respect to the vesicle trafficking of plants including rice, our understanding of the components and their coordinated regulation is very limited. At present, several vesicle trafficking apparatus components and cargo proteins have been identified and characterized in rice, but there still remain large unknowns concerning the organization and function of the rice vesicle trafficking system. In this review, we outline the main vesicle trafficking pathways of rice based on knowledge obtained in model organisms, and summarize current advances of rice vesicle trafficking. We also propose to develop methodologies applicable to rice and even other crops for further exploring the mysteries of vesicle trafficking in plants.
Collapse
Affiliation(s)
- Xiaobo Zhu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University at Wenjiang, Chengdu, Sichuan, China
| | - Junjie Yin
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University at Wenjiang, Chengdu, Sichuan, China
| | - Hongming Guo
- Environment-friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Yuping Wang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University at Wenjiang, Chengdu, Sichuan, China
| | - Bingtian Ma
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University at Wenjiang, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Tyagi S, Sarveswaran N, Higerd-Rusli GP, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Conserved but not critical: Trafficking and function of NaV1.7 are independent of highly conserved polybasic motifs. Front Mol Neurosci 2023; 16:1161028. [PMID: 37008789 PMCID: PMC10060856 DOI: 10.3389/fnmol.2023.1161028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Non-addictive treatment of chronic pain represents a major unmet clinical need. Peripheral voltage-gated sodium (NaV) channels are an attractive target for pain therapy because they initiate and propagate action potentials in primary afferents that detect and transduce noxious stimuli. NaV1.7 sets the gain on peripheral pain-signaling neurons and is the best validated peripheral ion channel involved in human pain, and previous work has shown that it is transported in vesicles in sensory axons which also carry Rab6a, a small GTPase known to be involved in vesicular packaging and axonal transport. Understanding the mechanism of the association between Rab6a and NaV1.7 could inform therapeutic modalities to decrease trafficking of NaV1.7 to the distal axonal membrane. Polybasic motifs (PBM) have been shown to regulate Rab-protein interactions in a variety of contexts. In this study, we explored whether two PBMs in the cytoplasmic loop that joins domains I and II of human NaV1.7 were responsible for association with Rab6a and regulate axonal trafficking of the channel. Using site-directed mutagenesis we generated NaV1.7 constructs with alanine substitutions in the two PBMs. Voltage-clamp recordings showed that the constructs retain wild-type like gating properties. Optical Pulse-chase Axonal Long-distance (OPAL) imaging in live sensory axons shows that mutations of these PBMs do not affect co-trafficking of Rab6a and NaV1.7, or the accumulation of the channel at the distal axonal surface. Thus, these polybasic motifs are not required for interaction of NaV1.7 with the Rab6a GTPase, or for trafficking of the channel to the plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Nivedita Sarveswaran
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Grant P. Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman,
| | - Sulayman D. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- Sulayman D. Dib-Hajj,
| |
Collapse
|
11
|
Abstract
Roseoloviruses (human herpesvirus 6A [HHV-6A], -6B, and -7) infect >90% of the human population during early childhood and are thought to remain latent or persistent throughout the life of the host. As such, these viruses are among the most pervasive and stealthy of all viruses; they must necessarily excel at escaping immune detection throughout the life of the host, and yet, very little is known about how these viruses so successfully escape host defenses. Here, we characterize the expression, trafficking, and posttranslational modifications of the HHV6B U20 gene product, which is encoded within a block of genes unique to the roseoloviruses. HHV-6B U20 trafficked slowly through the secretory system, receiving several posttranslational modifications to its N-linked glycans, indicative of surface-expressed glycoproteins, and eventually reaching the cell surface before being internalized. Interestingly, U20 is also phosphorylated on at least one Ser, Thr, or Tyr residue. These results provide a framework to understand the role(s) of U20 in evading host defenses. IMPORTANCE The roseolovirus U20 proteins are virus-encoded integral membrane glycoproteins possessing class I major histocompatibility complex (MHC)-like folds. Surprisingly, although U20 proteins from HHV-6A and -6B share 92% identity, recent studies ascribe different functions to HHV6A U20 and HHV6B U20. HHV6A U20 was shown to downregulate NKG2D ligands, while HHV6B U20 was shown to inhibit tumor necrosis factor alpha (TNF-α)-induced apoptosis during nonproductive infection with HHV6B (E. Kofod-Olsen, K. Ross-Hansen, M. H. Schleimann, D. K. Jensen, et al., J Virol 86:11483-11492, 2012, https://doi.org/10.1128/jvi.00847-12; A. E. Chaouat, B. Seliger, O. Mandelboim, D. Schmiedel, Front Immunol 12:714799, 2021, https://doi.org/10.3389/fimmu.2021.714799). Here, we have performed cell biological and biochemical characterization of the trafficking, glycosylation, and posttranslational modifications occurring on HHV6B U20.
Collapse
|
12
|
Harwood MC, Woo TT, Takeo Y, DiMaio D, Tsai B. HPV is a cargo for the COPI sorting complex during virus entry. SCIENCE ADVANCES 2023; 9:eadc9830. [PMID: 36662862 PMCID: PMC9858521 DOI: 10.1126/sciadv.adc9830] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/20/2022] [Indexed: 05/30/2023]
Abstract
During entry, human papillomavirus (HPV) traffics from the cell surface to the endosome and then to the trans-Golgi network (TGN) and Golgi apparatus. HPV must transit across the TGN/Golgi and exit these compartments to reach the nucleus to cause infection, although how these steps are accomplished is unclear. Combining cellular fractionation, unbiased proteomics, and gene knockdown strategies, we identified the coat protein complex I (COPI), a highly conserved protein complex that facilitates retrograde trafficking of cellular cargos, as a host factor required for HPV infection. Upon TGN/Golgi arrival, the cytoplasmic segment of HPV L2 binds directly to COPI. COPI depletion causes the accumulation of HPV in the TGN/Golgi, resembling the fate of a COPI binding-defective L2 mutant. We propose that the L2-COPI interaction drives HPV trafficking through the TGN and Golgi stacks during virus entry. This shows that an incoming virus is a cargo of the COPI complex.
Collapse
Affiliation(s)
- Mara C. Harwood
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tai-Ting Woo
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Yuka Takeo
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Daniel DiMaio
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Anders N, Wilson LFL, Sorieul M, Nikolovski N, Dupree P. β-1,4-Xylan backbone synthesis in higher plants: How complex can it be? FRONTIERS IN PLANT SCIENCE 2023; 13:1076298. [PMID: 36714768 PMCID: PMC9874913 DOI: 10.3389/fpls.2022.1076298] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
Xylan is a hemicellulose present in the cell walls of all land plants. Glycosyltransferases of the GT43 (IRX9/IRX9L and IRX14/IRX14L) and GT47 (IRX10/IRX10L) families are involved in the biosynthesis of its β-1,4-linked xylose backbone, which can be further modified by acetylation and sugar side chains. However, it remains unclear how the different enzymes work together to synthesize the xylan backbone. A xylan synthesis complex (XSC) has been described in the monocots wheat and asparagus, and co-expression of asparagus AoIRX9, AoIRX10 and AoIRX14A is required to form a catalytically active complex for secondary cell wall xylan biosynthesis. Here, we argue that an equivalent XSC exists for the synthesis of the primary cell wall of the eudicot Arabidopsis thaliana, consisting of IRX9L, IRX10L and IRX14. This would suggest the existence of distinct XSCs for primary and secondary cell wall xylan synthesis, reminiscent of the distinct cellulose synthesis complexes (CSCs) of the primary and secondary cell wall. In contrast to the CSC, in which each CESA protein has catalytic activity, the XSC seems to contain proteins with non-catalytic function with each component bearing potentially unique but crucial roles. Moreover, the core XSC formed by a combination of IRX9/IRX9L, IRX10/IRX10L and IRX14/IRX14L might not be stable in its composition during transit from the endoplasmic reticulum to the Golgi apparatus. Instead, potential dynamic changes of the XSC might be a means of regulating xylan biosynthesis to facilitate coordinated deposition of tailored polysaccharides in the plant cell wall.
Collapse
|
14
|
Winichayakul S, Curran A, Moraga R, Cookson R, Xue H, Crowther T, Roldan M, Bryan G, Roberts N. An alternative angiosperm DGAT1 topology and potential motifs in the N-terminus. FRONTIERS IN PLANT SCIENCE 2022; 13:951389. [PMID: 36186081 PMCID: PMC9523541 DOI: 10.3389/fpls.2022.951389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/05/2022] [Indexed: 06/16/2023]
Abstract
The highly variable cytoplasmic N-terminus of the plant diacylglycerol acyltransferase 1 (DGAT1) has been shown to have roles in oligomerization as well as allostery; however, the biological significance of the variation within this region is not understood. Comparing the coding sequences over the variable N-termini revealed the Poaceae DGAT1s contain relatively high GC compositional gradients as well as numerous direct and inverted repeats in this region. Using a variety of reciprocal chimeric DGAT1s from angiosperms we show that related N-termini had similar effects (positive or negative) on the accumulation of the recombinant protein in Saccharomyces cerevisiae. When expressed in Camelina sativa seeds the recombinant proteins of specific chimeras elevated total lipid content of the seeds as well as increased seed size. In addition, we combine N- and C-terminal as well as internal tags with high pH membrane reformation, protease protection and differential permeabilization. This led us to conclude the C-terminus is in the ER lumen; this contradicts earlier reports of the cytoplasmic location of plant DGAT1 C-termini.
Collapse
Affiliation(s)
- Somrutai Winichayakul
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Amy Curran
- ZeaKal Inc., San Diego, CA, United States
| | - Roger Moraga
- Bioinformatics and Statistics, AgResearch Ltd., Palmerston North, New Zealand
| | - Ruth Cookson
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Hong Xue
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Tracey Crowther
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Marissa Roldan
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
| | - Greg Bryan
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
- ZeaKal Inc., San Diego, CA, United States
| | - Nick Roberts
- Resilient Agriculture Innovation Centre of Excellence, AgResearch Ltd., Palmerston North, New Zealand
- ZeaKal Inc., San Diego, CA, United States
| |
Collapse
|
15
|
Ensinck MM, Carlon MS. One Size Does Not Fit All: The Past, Present and Future of Cystic Fibrosis Causal Therapies. Cells 2022; 11:cells11121868. [PMID: 35740997 PMCID: PMC9220995 DOI: 10.3390/cells11121868] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/04/2023] Open
Abstract
Cystic fibrosis (CF) is the most common monogenic disorder, caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Over the last 30 years, tremendous progress has been made in understanding the molecular basis of CF and the development of treatments that target the underlying defects in CF. Currently, a highly effective CFTR modulator treatment (Kalydeco™/Trikafta™) is available for 90% of people with CF. In this review, we will give an extensive overview of past and ongoing efforts in the development of therapies targeting the molecular defects in CF. We will discuss strategies targeting the CFTR protein (i.e., CFTR modulators such as correctors and potentiators), its cellular environment (i.e., proteostasis modulation, stabilization at the plasma membrane), the CFTR mRNA (i.e., amplifiers, nonsense mediated mRNA decay suppressors, translational readthrough inducing drugs) or the CFTR gene (gene therapies). Finally, we will focus on how these efforts can be applied to the 15% of people with CF for whom no causal therapy is available yet.
Collapse
Affiliation(s)
- Marjolein M. Ensinck
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Flanders, Belgium;
| | - Marianne S. Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Flanders, Belgium;
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Flanders, Belgium
- Correspondence:
| |
Collapse
|
16
|
Hatanaka R, Araki E, Hane M, Go S, Wu D, Kitajima K, Sato C. The α2,8-sialyltransferase 6 (St8sia6) localizes in the ER and enhances the anchorage-independent cell growth in cancer. Biochem Biophys Res Commun 2022; 608:52-58. [PMID: 35390672 DOI: 10.1016/j.bbrc.2022.03.146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/27/2022] [Indexed: 01/06/2023]
Abstract
Sialylation, the final stage of post-translational modification of proteins, is achieved in the Golgi apparatus and is related to the malignant phenotype of cancer. Disialylation of ganglioside (GD3) by St8sia1 and polysialylation by St8sia2 and 4 have been shown to be related to malignant phenotypes; however, di/oligosialylation by St8sia6 is still unknown. In this study, we analyzed the malignant phenotype of St8sia6 and found that upregulation of St8sia6 in melanoma B16 cells increased anchorage-independent cell growth, which was not due to sialic acid cleavage by a sialidase. Moreover, unlike other sialyltransferases, St8sia6 localized to the endoplasmic reticulum (ER). We found that the localization to the Golgi apparatus could be regulated by swapping experiments using St8sia2; however, the malignant phenotype did not change. These data demonstrate that the enhancement of anchorage-independent cell growth by St8sia6 is not due to its localization of ER, but is due to the expression of the protein itself.
Collapse
Affiliation(s)
- Rina Hatanaka
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Erino Araki
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Shiori Go
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Di Wu
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Integrated Glyco-Biomedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa, Nagoya, 464-8601, Japan.
| |
Collapse
|
17
|
Matsuoka K, Imahashi N, Ohno M, Ode H, Nakata Y, Kubota M, Sugimoto A, Imahashi M, Yokomaku Y, Iwatani Y. SARS-CoV-2 accessory protein ORF8 is secreted extracellularly as a glycoprotein homodimer. J Biol Chem 2022; 298:101724. [PMID: 35157849 PMCID: PMC8832879 DOI: 10.1016/j.jbc.2022.101724] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 01/03/2023] Open
Abstract
ORF8 is an accessory protein encoded by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Consensus regarding the biological functions of ORF8 is lacking, largely because the fundamental characteristics of this protein in cells have not been determined. To clarify these features, we herein established an ORF8 expression system in 293T cells. Using this system, approximately 41% of the ORF8 expressed in 293T cells were secreted extracellularly as a glycoprotein homodimer with inter/intramolecular disulfide bonds. Intracellular ORF8 was sensitive to the glycosidase Endo H, whereas the secreted portion was Endo-H-resistant, suggesting that secretion occurs via a conventional pathway. Additionally, immunoblotting analysis showed that the total amounts of the major histocompatibility complex class Ι (MHC-I), angiotensin-converting enzyme 2 (ACE2), and SARS-CoV-2 spike (CoV-2 S) proteins coexpressed in cells were not changed by the increased ORF8 expression, although FACS analysis revealed that the expression of the cell surface MHC-I protein, but not that of ACE2 and CoV-2 S proteins, was reduced by ORF8 expression. Finally, we demonstrate by RNA-seq analysis that ORF8 had no significant stimulatory effects in human primary monocyte-derived macrophages (MDMs). Taken together, our results provide fundamental evidence that the ORF8 glycoprotein acts as a secreted homodimer, and its functions are likely associated with the intracellular transport and/or extracellular signaling in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kazuhiro Matsuoka
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Nobuhiko Imahashi
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Miki Ohno
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Hirotaka Ode
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Yoshihiro Nakata
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan; Department of AIDS Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mai Kubota
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Atsuko Sugimoto
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Mayumi Imahashi
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Yoshiyuki Yokomaku
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Yasumasa Iwatani
- Department of Infectious Diseases and Immunology, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan; Department of AIDS Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| |
Collapse
|
18
|
Virolainen MS, Søltoft CL, Pedersen PA, Ellgaard L. Production of an Active, Human Membrane Protein in Saccharomyces cerevisiae: Full-Length FICD. Int J Mol Sci 2022; 23:ijms23052458. [PMID: 35269596 PMCID: PMC8910494 DOI: 10.3390/ijms23052458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/10/2022] Open
Abstract
The human Fic domain-containing protein (FICD) is a type II endoplasmic reticulum (ER) membrane protein that is important for the maintenance of ER proteostasis. Structural and in vitro biochemical characterisation of FICD AMPylase and deAMPylase activity have been restricted to the soluble ER-luminal domain produced in Escherichia coli. Information about potentially important features, such as structural motifs, modulator binding sites or other regulatory elements, is therefore missing for the approximately 100 N-terminal residues including the transmembrane region of FICD. Expressing and purifying the required quantity and quality of membrane proteins is demanding because of the low yields and poor stability often observed. Here, we produce full-length FICD by combining a Saccharomyces cerevisiae-based platform with green fluorescent protein (GFP) tagging to optimise the conditions for expression, solubilisation and purification. We subsequently employ these conditions to purify milligram quantities of His-tagged FICD per litre of culture, and show that the purified, detergent-solubilised membrane protein is an active deAMPylating enzyme. Our work provides a straightforward methodology for producing not only full-length FICD, but also other membrane proteins in S. cerevisiae for structural and biochemical characterisation.
Collapse
Affiliation(s)
- Minttu S. Virolainen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark; (M.S.V.); (C.L.S.)
| | - Cecilie L. Søltoft
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark; (M.S.V.); (C.L.S.)
| | - Per A. Pedersen
- Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2200 Copenhagen, Denmark
- Correspondence: (P.A.P.); (L.E.)
| | - Lars Ellgaard
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark; (M.S.V.); (C.L.S.)
- Correspondence: (P.A.P.); (L.E.)
| |
Collapse
|
19
|
D’Souza Z, Sumya FT, Khakurel A, Lupashin V. Getting Sugar Coating Right! The Role of the Golgi Trafficking Machinery in Glycosylation. Cells 2021; 10:cells10123275. [PMID: 34943782 PMCID: PMC8699264 DOI: 10.3390/cells10123275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022] Open
Abstract
The Golgi is the central organelle of the secretory pathway and it houses the majority of the glycosylation machinery, which includes glycosylation enzymes and sugar transporters. Correct compartmentalization of the glycosylation machinery is achieved by retrograde vesicular trafficking as the secretory cargo moves forward by cisternal maturation. The vesicular trafficking machinery which includes vesicular coats, small GTPases, tethers and SNAREs, play a major role in coordinating the Golgi trafficking thereby achieving Golgi homeostasis. Glycosylation is a template-independent process, so its fidelity heavily relies on appropriate localization of the glycosylation machinery and Golgi homeostasis. Mutations in the glycosylation enzymes, sugar transporters, Golgi ion channels and several vesicle tethering factors cause congenital disorders of glycosylation (CDG) which encompass a group of multisystem disorders with varying severities. Here, we focus on the Golgi vesicle tethering and fusion machinery, namely, multisubunit tethering complexes and SNAREs and their role in Golgi trafficking and glycosylation. This review is a comprehensive summary of all the identified CDG causing mutations of the Golgi trafficking machinery in humans.
Collapse
|
20
|
Lujan P, Campelo F. Should I stay or should I go? Golgi membrane spatial organization for protein sorting and retention. Arch Biochem Biophys 2021; 707:108921. [PMID: 34038703 DOI: 10.1016/j.abb.2021.108921] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/12/2021] [Accepted: 05/03/2021] [Indexed: 12/23/2022]
Abstract
The Golgi complex is the membrane-bound organelle that lies at the center of the secretory pathway. Its main functions are to maintain cellular lipid homeostasis, to orchestrate protein processing and maturation, and to mediate protein sorting and export. These functions are not independent of one another, and they all require that the membranes of the Golgi complex have a well-defined biochemical composition. Importantly, a finely-regulated spatiotemporal organization of the Golgi membrane components is essential for the correct performance of the organelle. In here, we review our current mechanistic and molecular understanding of how Golgi membranes are spatially organized in the lateral and axial directions to fulfill their functions. In particular, we highlight the current evidence and proposed models of intra-Golgi transport, as well as the known mechanisms for the retention of Golgi residents and for the sorting and export of transmembrane cargo proteins. Despite the controversies, conflicting evidence, clashes between models, and technical limitations, the field has moved forward and we have gained extensive knowledge in this fascinating topic. However, there are still many important questions that remain to be completely answered. We hope that this review will help boost future investigations on these issues.
Collapse
Affiliation(s)
- Pablo Lujan
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Barcelona, Spain.
| | - Felix Campelo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Barcelona, Spain.
| |
Collapse
|
21
|
Wouters R, Michiels C, Sannerud R, Kleizen B, Dillen K, Vermeire W, Ayala AE, Demedts D, Schekman R, Annaert W. Assembly of γ-secretase occurs through stable dimers after exit from the endoplasmic reticulum. J Cell Biol 2021; 220:212501. [PMID: 34292306 PMCID: PMC8302450 DOI: 10.1083/jcb.201911104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/29/2020] [Accepted: 06/07/2021] [Indexed: 01/22/2023] Open
Abstract
γ-Secretase affects many physiological processes through targeting >100 substrates; malfunctioning links γ-secretase to cancer and Alzheimer’s disease. The spatiotemporal regulation of its stoichiometric assembly remains unresolved. Fractionation, biochemical assays, and imaging support prior formation of stable dimers in the ER, which, after ER exit, assemble into full complexes. In vitro ER budding shows that none of the subunits is required for the exit of others. However, knockout of any subunit leads to the accumulation of incomplete subcomplexes in COPII vesicles. Mutating a DPE motif in presenilin 1 (PSEN1) abrogates ER exit of PSEN1 and PEN-2 but not nicastrin. We explain this by the preferential sorting of PSEN1 and nicastrin through Sec24A and Sec24C/D, respectively, arguing against full assembly before ER exit. Thus, dimeric subcomplexes aided by Sec24 paralog selectivity support a stepwise assembly of γ-secretase, controlling final levels in post-Golgi compartments.
Collapse
Affiliation(s)
- Rosanne Wouters
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Christine Michiels
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ragna Sannerud
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bertrand Kleizen
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Katleen Dillen
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Wendy Vermeire
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Abril Escamilla Ayala
- Vlaams Instituut voor Biotechnologie BioImaging Core, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Leuven, Belgium
| | - David Demedts
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Randy Schekman
- Department of Molecular and Cell Biology and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA
| | - Wim Annaert
- Laboratory for Membrane Trafficking, Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Chau S, Fujii A, Wang Y, Vandebroek A, Goda W, Yasui M, Abe Y. Di-lysine motif-like sequences formed by deleting the C-terminal domain of aquaporin-4 prevent its trafficking to the plasma membrane. Genes Cells 2021; 26:152-164. [PMID: 33474763 DOI: 10.1111/gtc.12829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 11/30/2022]
Abstract
Aquaporin-4 is a transmembrane water channel protein, the C-terminal domain of which is facing the cytosol. In the process of investigating the role of the C-terminal domain of aquaporin-4 with regard to intracellular trafficking, we observed that a derivative of aquaporin-4, in which the C-terminal 53 amino acids had been removed (Δ271-323), was localized to intracellular compartments, including the endoplasmic reticulum, but was not expressed on the plasma membranes. This was determined by immunofluorescence staining and labeling of the cells with monoclonal antibody specifically recognizing the extracellular domain of aquaporin-4, followed by confocal microscopy and flow cytometry. Deletion of additional amino acids in the C-terminal domain of aquaporin-4 led to its redistribution to the plasma membrane. This suggests that the effect of the 53-amino acid deletion on the subcellular localization of aquaporin-4 could be attributed to the formation of a signal at the C terminus that retained aquaporin-4 in intracellular compartments, rather than the loss of a signal required for plasma membrane targeting. Substitution of the lysine at position 268 with alanine could rescue the Δ271-323-associated retention in the cytosol, suggesting that the C-terminal sequence of the mutant served as a signal similar to a di-lysine motif.
Collapse
Affiliation(s)
- Simon Chau
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Fujii
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Yingqi Wang
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Arno Vandebroek
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Wakami Goda
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.,Center for Water Biology & Medicine, Keio University Global Research Institute, Tokyo, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.,Center for Water Biology & Medicine, Keio University Global Research Institute, Tokyo, Japan
| |
Collapse
|
23
|
Brahimi F, Galan A, Jmaeff S, Barcelona PF, De Jay N, Dejgaard K, Young JC, Kleinman CL, Thomas DY, Saragovi HU. Alternative Splicing of a Receptor Intracellular Domain Yields Different Ectodomain Conformations, Enabling Isoform-Selective Functional Ligands. iScience 2020; 23:101447. [PMID: 32829283 PMCID: PMC7452315 DOI: 10.1016/j.isci.2020.101447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/13/2020] [Accepted: 08/06/2020] [Indexed: 01/04/2023] Open
Abstract
Events at a receptor ectodomain affect the intracellular domain conformation, activating signal transduction (out-to-in conformational effects). We investigated the reverse direction (in-to-out) where the intracellular domain may impact on ectodomain conformation. The primary sequences of naturally occurring TrkC receptor isoforms (TrkC-FL and TrkC.T1) only differ at the intracellular domain. However, owing to their differential association with Protein Disulfide Isomerase the isoforms have different disulfide bonding and conformations at the ectodomain. Conformations were exploited to develop artificial ligands, mAbs, and small molecules, with isoform-specific binding and biased activation. Consistent, the physiological ligands NT-3 and PTP-sigma bind both isoforms, but NT-3 activates all signaling pathways, whereas PTP-sigma activates biased signals. Our data support an "in-to-out" model controlling receptor ectodomain conformation, a strategy that enables heterogeneity in receptors, ligands, and bioactivity. These concepts may be extended to the many wild-type or oncogenic receptors with known isoforms.
Collapse
Affiliation(s)
- Fouad Brahimi
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Alba Galan
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Sean Jmaeff
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Pharmacology, McGill University, Montreal, QC, Canada
| | - Pablo F. Barcelona
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Nicolas De Jay
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Kurt Dejgaard
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Jason C. Young
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Claudia L. Kleinman
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - David Y. Thomas
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - H. Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Pharmacology, McGill University, Montreal, QC, Canada
- Department of Ophthalmology and Visual Science, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Menzel J, Kownatzki-Danger D, Tokar S, Ballone A, Unthan-Fechner K, Kilisch M, Lenz C, Urlaub H, Mori M, Ottmann C, Shattock MJ, Lehnart SE, Schwappach B. 14-3-3 binding creates a memory of kinase action by stabilizing the modified state of phospholamban. Sci Signal 2020; 13:13/647/eaaz1436. [DOI: 10.1126/scisignal.aaz1436] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cardiac membrane protein phospholamban (PLN) is targeted by protein kinase A (PKA) at Ser16and by Ca2+/calmodulin-dependent protein kinase II (CaMKII) at Thr17. β-Adrenergic stimulation and PKA-dependent phosphorylation of Ser16acutely stimulate the sarcoplasmic reticulum calcium pump (SERCA) by relieving its inhibition by PLN. CaMKII-dependent phosphorylation may lead to longer-lasting SERCA stimulation and may sustain maladaptive Ca2+handling. Here, we demonstrated that phosphorylation at either Ser16or Thr17converted PLN into a target for the phosphoadaptor protein 14-3-3 with different affinities. 14-3-3 proteins were localized within nanometers of PLN and endogenous 14-3-3 coimmunoprecipitated with pentameric PLN from cardiac membranes. Molecular dynamics simulations predicted different molecular contacts for peptides phosphorylated at Ser16or Thr17with the binding groove of 14-3-3, resulting in varied binding affinities. 14-3-3 binding protected either PLN phosphosite from dephosphorylation. β-Adrenergic stimulation of isolated adult cardiomyocytes resulted in the membrane recruitment of endogenous 14-3-3. The exogenous addition of 14-3-3 to β-adrenergic–stimulated cardiomyocytes led to prolonged SERCA activation, presumably because 14-3-3 protected PLN pentamers from dephosphorylation. Phosphorylation of Ser16was disrupted by the cardiomyopathy-associated ∆Arg14mutation, implying that phosphorylation of Thr17by CaMKII may become crucial for 14-3-3 recruitment to ∆Arg14PLN. Consistent with PLN acting as a dynamic hub in the control of Ca2+handling, our results identify 14-3-3 binding to PLN as a contractility-augmenting mechanism.
Collapse
Affiliation(s)
- Julia Menzel
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Daniel Kownatzki-Danger
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, Universitätsmedizin Göttingen, Robert-Koch-Straße 42a, 37075 Göttingen, Germany
| | - Sergiy Tokar
- School of Cardiovascular Medicine and Sciences, King’s College London, Westminster Bridge Road, London SE17H, UK
| | - Alice Ballone
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P. O. Box 513, 5600MB Eindhoven, Netherlands
| | - Kirsten Unthan-Fechner
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Markus Kilisch
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Christof Lenz
- Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- Max-Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
| | - Henning Urlaub
- Bioanalytics Group, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- Max-Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P. O. Box 513, 5600MB Eindhoven, Netherlands
| | - Michael J. Shattock
- School of Cardiovascular Medicine and Sciences, King’s College London, Westminster Bridge Road, London SE17H, UK
| | - Stephan E. Lehnart
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, Universitätsmedizin Göttingen, Robert-Koch-Straße 42a, 37075 Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC), University of Goettingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
- Max-Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC), University of Goettingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| |
Collapse
|
25
|
Annaert W, Kaether C. Bring it back, bring it back, don't take it away from me - the sorting receptor RER1. J Cell Sci 2020; 133:133/17/jcs231423. [PMID: 32873699 DOI: 10.1242/jcs.231423] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The quote "bring it back, bring it back, don't take it away from me" from Queen's Love of my life describes the function of the sorting receptor RER1, a 23 kDa protein with four transmembrane domains (TMDs) that localizes to the intermediate compartment and the cis-Golgi. From there it returns escaped proteins that are not supposed to leave the endoplasmic reticulum (ER) back to it. Unique about RER1 is its ability to recognize its ligands through binding motifs in TMDs. Among its substrates are ER-resident proteins, as well as unassembled subunits of multimeric complexes that are retrieved back into the ER, this way guarding the full assembly of their respective complexes. The basic mechanisms for RER1-dependent retrieval have been already elucidated some years ago in yeast. More recently, several important cargoes of RER1 have been described in mammalian cells, and the in vivo role of RER1 is being unveiled by using mouse models. In this Review, we give an overview of the cell biology of RER1 in different models, discuss its controversial role in the brain and provide an outlook on future directions for RER1 research.
Collapse
Affiliation(s)
- Wim Annaert
- VIB Center for Brain and Disease Research & KU Leuven, Department of Neurosciences, Gasthuisberg, B-3000 Leuven, Belgium
| | - Christoph Kaether
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07745 Jena, Germany
| |
Collapse
|
26
|
Nie Y, Dai B, Guo X, Liu D. Cleavage of FNDC5 and insights into its maturation process. Mol Cell Endocrinol 2020; 510:110840. [PMID: 32360564 DOI: 10.1016/j.mce.2020.110840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/02/2023]
Abstract
FNDC5 corresponds to an irisin precursor that increases with exercise. Studies suggest that irisin mediates beneficial effects in adipose tissues, skeletal muscle, bone, and brain. However, the cleavage and maturation processes of FNDC5 have not been clearly identified. This study aimed to show that the signal peptide and transmembrane domain of FNDC5 were associated with the secretion of its ectodomain. Localization studies identified the signal peptide that was responsible for endoplasmic reticulum targeting activity of nascent FNDC5 and showed that the FNDC5 ectodomain corresponding to irisin could be transported across the membrane by a transmembrane domain. Analysis of cleavage constructs revealed that the ectodomain of FNDC5 could be cleaved from its signal peptide and transmembrane attachment. Genetic ablation of the signal peptide cleavage site blocked N-glycosylation of FNDC5. Identification of the FNDC5 maturation process should facilitate our understanding of irisin secretion.
Collapse
Affiliation(s)
- Yongwei Nie
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, 24 Zhaojun Road, Hohhot, 010070, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Bai Dai
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, 24 Zhaojun Road, Hohhot, 010070, China
| | - Xudong Guo
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, 24 Zhaojun Road, Hohhot, 010070, China.
| | - Dongjun Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, 24 Zhaojun Road, Hohhot, 010070, China.
| |
Collapse
|
27
|
Lara R, Adinolfi E, Harwood CA, Philpott M, Barden JA, Di Virgilio F, McNulty S. P2X7 in Cancer: From Molecular Mechanisms to Therapeutics. Front Pharmacol 2020; 11:793. [PMID: 32581786 PMCID: PMC7287489 DOI: 10.3389/fphar.2020.00793] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
P2X7 is a transmembrane receptor expressed in multiple cell types including neurons, dendritic cells, macrophages, monocytes, B and T cells where it can drive a wide range of physiological responses from pain transduction to immune response. Upon activation by its main ligand, extracellular ATP, P2X7 can form a nonselective channel for cations to enter the cell. Prolonged activation of P2X7, via high levels of extracellular ATP over an extended time period can lead to the formation of a macropore, leading to depolarization of the plasma membrane and ultimately to cell death. Thus, dependent on its activation state, P2X7 can either drive cell survival and proliferation, or induce cell death. In cancer, P2X7 has been shown to have a broad range of functions, including playing key roles in the development and spread of tumor cells. It is therefore unsurprising that P2X7 has been reported to be upregulated in several malignancies. Critically, ATP is present at high extracellular concentrations in the tumor microenvironment (TME) compared to levels observed in normal tissues. These high levels of ATP should present a survival challenge for cancer cells, potentially leading to constitutive receptor activation, prolonged macropore formation and ultimately to cell death. Therefore, to deliver the proven advantages for P2X7 in driving tumor survival and metastatic potential, the P2X7 macropore must be tightly controlled while retaining other functions. Studies have shown that commonly expressed P2X7 splice variants, distinct SNPs and post-translational receptor modifications can impair the capacity of P2X7 to open the macropore. These receptor modifications and potentially others may ultimately protect cancer cells from the negative consequences associated with constitutive activation of P2X7. Significantly, the effects of both P2X7 agonists and antagonists in preclinical tumor models of cancer demonstrate the potential for agents modifying P2X7 function, to provide innovative cancer therapies. This review summarizes recent advances in understanding of the structure and functions of P2X7 and how these impact P2X7 roles in cancer progression. We also review potential therapeutic approaches directed against P2X7.
Collapse
Affiliation(s)
- Romain Lara
- Biosceptre (UK) Limited, Cambridge, United Kingdom
| | - Elena Adinolfi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mike Philpott
- Centre for Cutaneous Research, Blizard Institute, Bart's & The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
28
|
Weinert S, Gimber N, Deuschel D, Stuhlmann T, Puchkov D, Farsi Z, Ludwig CF, Novarino G, López-Cayuqueo KI, Planells-Cases R, Jentsch TJ. Uncoupling endosomal CLC chloride/proton exchange causes severe neurodegeneration. EMBO J 2020; 39:e103358. [PMID: 32118314 PMCID: PMC7196918 DOI: 10.15252/embj.2019103358] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/02/2023] Open
Abstract
CLC chloride/proton exchangers may support acidification of endolysosomes and raise their luminal Cl− concentration. Disruption of endosomal ClC‐3 causes severe neurodegeneration. To assess the importance of ClC‐3 Cl−/H+ exchange, we now generate Clcn3unc/unc mice in which ClC‐3 is converted into a Cl− channel. Unlike Clcn3−/− mice, Clcn3unc/unc mice appear normal owing to compensation by ClC‐4 with which ClC‐3 forms heteromers. ClC‐4 protein levels are strongly reduced in Clcn3−/−, but not in Clcn3unc/unc mice because ClC‐3unc binds and stabilizes ClC‐4 like wild‐type ClC‐3. Although mice lacking ClC‐4 appear healthy, its absence in Clcn3unc/unc/Clcn4−/− mice entails even stronger neurodegeneration than observed in Clcn3−/− mice. A fraction of ClC‐3 is found on synaptic vesicles, but miniature postsynaptic currents and synaptic vesicle acidification are not affected in Clcn3unc/unc or Clcn3−/− mice before neurodegeneration sets in. Both, Cl−/H+‐exchange activity and the stabilizing effect on ClC‐4, are central to the biological function of ClC‐3.
Collapse
Affiliation(s)
- Stefanie Weinert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Niclas Gimber
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Dorothea Deuschel
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Till Stuhlmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Dmytro Puchkov
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Zohreh Farsi
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Carmen F Ludwig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Gaia Novarino
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Karen I López-Cayuqueo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Rosa Planells-Cases
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
29
|
Yano H, Liu L, Naing S, Shi L. The Effects of Terminal Tagging on Homomeric Interactions of the Sigma 1 Receptor. Front Neurosci 2019; 13:1356. [PMID: 31920515 PMCID: PMC6930887 DOI: 10.3389/fnins.2019.01356] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/02/2019] [Indexed: 11/13/2022] Open
Abstract
The sigma 1 receptor (σ1R) has been implicated in cancers, neurological disorders, and substance use disorders. Yet, its molecular and cellular functions have not been well-understood. Recent crystal structures of σ1R reveal a single N-terminal transmembrane segment and C-terminal ligand-binding domain, and a trimeric organization. Nevertheless, outstanding issues surrounding the functional or pharmacological relevance of σ1R oligomerization remain, such as the minimal protomeric unit and the differentially altered oligomerization states by different classes of ligands. Western blot (WB) assays have been widely used to investigate protein oligomerizations. However, the unique topology of σ1R renders several intertwined challenges in WB. Here we describe a WB protocol without temperature denaturization to study the ligand binding effects on the oligomerization state of σ1R. Using this approach, we observed unexpected ladder-like incremental migration pattern of σ1R, demonstrating preserved homomeric interactions in the detergent environment. We compared the migration patterns of intact σ1R construct and the C-terminally tagged σ1R constructs, and found similar trends in response to drug treatments. In contrast, N-terminally tagged σ1R constructs show opposite trends to that of the intact construct, suggesting distorted elicitation of the ligand binding effects on oligomerization. Together, our findings indicate that the N-terminus plays an important role in eliciting the impacts of bound ligands, whereas the C-terminus is amenable for modifications for biochemical studies.
Collapse
Affiliation(s)
- Hideaki Yano
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Leanne Liu
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Sett Naing
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
30
|
A novel ER membrane protein Ehg1/May24 plays a critical role in maintaining multiple nutrient permeases in yeast under high-pressure perturbation. Sci Rep 2019; 9:18341. [PMID: 31797992 PMCID: PMC6892922 DOI: 10.1038/s41598-019-54925-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022] Open
Abstract
Previously, we isolated 84 deletion mutants in Saccharomyces cerevisiae auxotrophic background that exhibited hypersensitive growth under high hydrostatic pressure and/or low temperature. Here, we observed that 24 deletion mutants were rescued by the introduction of four plasmids (LEU2, HIS3, LYS2, and URA3) together to grow at 25 MPa, thereby suggesting close links between the genes and nutrient uptake. Most of the highly ranked genes were poorly characterized, including MAY24/YPR153W. May24 appeared to be localized in the endoplasmic reticulum (ER) membrane. Therefore, we designated this gene as EHG (ER-associated high-pressure growth gene) 1. Deletion of EHG1 led to reduced nutrient transport rates and decreases in the nutrient permease levels at 25 MPa. These results suggest that Ehg1 is required for the stability and functionality of the permeases under high pressure. Ehg1 physically interacted with nutrient permeases Hip1, Bap2, and Fur4; however, alanine substitutions for Pro17, Phe19, and Pro20, which were highly conserved among Ehg1 homologues in various yeast species, eliminated interactions with the permeases as well as the high-pressure growth ability. By functioning as a novel chaperone that facilitated coping with high-pressure-induced perturbations, Ehg1 could exert a stabilizing effect on nutrient permeases when they are present in the ER.
Collapse
|
31
|
Gpr63 is a modifier of microcephaly in Ttc21b mouse mutants. PLoS Genet 2019; 15:e1008467. [PMID: 31730647 PMCID: PMC6881074 DOI: 10.1371/journal.pgen.1008467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/27/2019] [Accepted: 10/08/2019] [Indexed: 11/19/2022] Open
Abstract
The primary cilium is a signaling center critical for proper embryonic development. Previous studies have demonstrated that mice lacking Ttc21b have impaired retrograde trafficking within the cilium and multiple organogenesis phenotypes, including microcephaly. Interestingly, the severity of the microcephaly in Ttc21baln/aln homozygous null mutants is considerably affected by the genetic background and mutants on an FVB/NJ (FVB) background develop a forebrain significantly smaller than mutants on a C57BL/6J (B6) background. We performed a Quantitative Trait Locus (QTL) analysis to identify potential genetic modifiers and identified two regions linked to differential forebrain size: modifier of alien QTL1 (Moaq1) on chromosome 4 at 27.8 Mb and Moaq2 on chromosome 6 at 93.6 Mb. These QTLs were validated by constructing congenic strains. Further analysis of Moaq1 identified an orphan G-protein coupled receptor (GPCR), Gpr63, as a candidate gene. We identified a SNP that is polymorphic between the FVB and B6 strains in Gpr63 and creates a missense mutation predicted to be deleterious in the FVB protein. We used CRISPR-Cas9 genome editing to create two lines of FVB congenic mice: one with the B6 sequence of Gpr63 and the other with a deletion allele leading to a truncation of the GPR63 C-terminal tail. We then demonstrated that Gpr63 can localize to the cilium in vitro. These alleles affect ciliary localization of GPR63 in vitro and genetically interact with Ttc21baln/aln as Gpr63;Ttc21b double mutants show unique phenotypes including spina bifida aperta and earlier embryonic lethality. This validated Gpr63 as a modifier of multiple Ttc21b neural phenotypes and strongly supports Gpr63 as a causal gene (i.e., a quantitative trait gene, QTG) within the Moaq1 QTL. TTC21B in humans is a known ciliopathy gene and contributes to the pathophysiology of a number of ciliopathies. Mice homozygous for a null allele of Ttc21b also have a spectrum of ciliopathy phenotypes, including microcephaly (small brain). Further work has shown that the severity of the microcephaly significantly depends on the genetic background of the mouse model. The genetic mechanisms of the Ttc21b pathophysiology and the interacting gene network remain far from understood. As an initial attempt to understand the underlying mechanism(s) underlying the variable effects on brain size, we performed a quantitative trait locus (QTL) analysis and found two regions of genomic significance that correlated with smaller brain size. We confirmed both QTLs with congenic lines. One of the two regions was small enough that we considered candidate genes and hypothesized Gpr63 might be a contributing locus for a number of reasons. We evaluated this hypothesis directly with precise variant creation using genome editing and provide evidence that Ttc21b and Gpr63 do indeed genetically interact. Thus, we have been able to combine classical QTL analysis and genome editing to directly test the resulting hypothesis.
Collapse
|
32
|
Jiménez-González V, Ogalla-García E, García-Quintanilla M, García-Quintanilla A. Deciphering GRINA/Lifeguard1: Nuclear Location, Ca 2+ Homeostasis and Vesicle Transport. Int J Mol Sci 2019; 20:ijms20164005. [PMID: 31426446 PMCID: PMC6719933 DOI: 10.3390/ijms20164005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023] Open
Abstract
The Glutamate Receptor Ionotropic NMDA-Associated Protein 1 (GRINA) belongs to the Lifeguard family and is involved in calcium homeostasis, which governs key processes, such as cell survival or the release of neurotransmitters. GRINA is mainly associated with membranes of the endoplasmic reticulum, Golgi, endosome, and the cell surface, but its presence in the nucleus has not been explained yet. Here we dissect, with the help of different software tools, the potential roles of GRINA in the cell and how they may be altered in diseases, such as schizophrenia or celiac disease. We describe for the first time that the cytoplasmic N-terminal half of GRINA (which spans a Proline-rich domain) contains a potential DNA-binding sequence, in addition to cleavage target sites and probable PY-nuclear localization sequences, that may enable it to be released from the rest of the protein and enter the nucleus under suitable conditions, where it could participate in the transcription, alternative splicing, and mRNA export of a subset of genes likely involved in lipid and sterol synthesis, ribosome biogenesis, or cell cycle progression. To support these findings, we include additional evidence based on an exhaustive review of the literature and our preliminary data of the protein–protein interaction network of GRINA.
Collapse
Affiliation(s)
| | - Elena Ogalla-García
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Meritxell García-Quintanilla
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Albert García-Quintanilla
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
33
|
Welch LG, Munro S. A tale of short tails, through thick and thin: investigating the sorting mechanisms of Golgi enzymes. FEBS Lett 2019; 593:2452-2465. [DOI: 10.1002/1873-3468.13553] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Lawrence G. Welch
- MRC Laboratory of Molecular Biology Francis Crick Avenue Cambridge UK
| | - Sean Munro
- MRC Laboratory of Molecular Biology Francis Crick Avenue Cambridge UK
| |
Collapse
|
34
|
α7 nicotinic acetylcholine receptor upregulation by anti-apoptotic Bcl-2 proteins. Nat Commun 2019; 10:2746. [PMID: 31227712 PMCID: PMC6588605 DOI: 10.1038/s41467-019-10723-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/24/2019] [Indexed: 01/07/2023] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) mediate and modulate synaptic transmission throughout the brain, and contribute to learning, memory, and behavior. Dysregulation of α7-type nAChRs in neuropsychiatric as well as immunological and oncological diseases makes them attractive targets for pharmaceutical development. Recently, we identified NACHO as an essential chaperone for α7 nAChRs. Leveraging the robust recombinant expression of α7 nAChRs with NACHO, we utilized genome-wide cDNA library screening and discovered that several anti-apoptotic Bcl-2 family proteins further upregulate receptor assembly and cell surface expression. These effects are mediated by an intracellular motif on α7 that resembles the BH3 binding domain of pro-apoptotic Bcl-2 proteins, and can be blocked by BH3 mimetic Bcl-2 inhibitors. Overexpression of Bcl-2 member Mcl-1 in neurons enhanced surface expression of endogenous α7 nAChRs, while a combination of chemotherapeutic Bcl2-inhibitors suppressed neuronal α7 receptor assembly. These results demonstrate that Bcl-2 proteins link α7 nAChR assembly to cell survival pathways. The α7 nicotinic acetylcholine receptor (nAChR) plays a major role in shaping the activity of neuronal circuits and contributes to the pathophysiology of several neurological disorders. Following cDNA library screening, the authors identify anti-apoptotic, Bcl-2 family proteins as enhancers of α7 nAChR assembly, acting through an intracellular BH3-like domain during receptor biogenesis in the endoplasmic reticulum.
Collapse
|
35
|
Joyce S, Nour AM. Blocking transmembrane219 protein signaling inhibits autophagy and restores normal cell death. PLoS One 2019; 14:e0218091. [PMID: 31220095 PMCID: PMC6586287 DOI: 10.1371/journal.pone.0218091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/24/2019] [Indexed: 02/04/2023] Open
Abstract
Autophagy plays a vital role in tumor therapy and survival of dormant tumor cells. Here we describe a novel function of a protein known as Transmembrane 219 (TM219) as an autophagy activator. TM219 is a small membrane protein expressed in all known human tissues except the thymus. We used biochemical approaches to identify calmodulin and calmodulin dependent protein kinase II as a part of TM219 protein complex. Then, we employed in vitro reconstitution system and fluorescence anisotropy to study the requirements of TM219 to bind calmodulin in vitro. We also used this system to study the effects of a synthetic peptide derived from the sequence of the short cytoplasmic tail of TM219 (SCTT) on calmodulin-TM219 receptor interactions. We conjugated SCTT peptide with a pH Low Insertion peptide (pHLIP) for optimal cellular delivery. We finally tested the effects of SCTT-pHLIP on triple negative human breast cancer cells in three dimension culture. Our data defined a novel function of TM219 protein and an efficient approach to inhibit it.
Collapse
Affiliation(s)
- Sean Joyce
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Adel M. Nour
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
36
|
Shiwarski DJ, Crilly SE, Dates A, Puthenveedu MA. Dual RXR motifs regulate nerve growth factor-mediated intracellular retention of the delta opioid receptor. Mol Biol Cell 2019; 30:680-690. [PMID: 30601694 PMCID: PMC6589700 DOI: 10.1091/mbc.e18-05-0292] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/21/2018] [Accepted: 12/27/2018] [Indexed: 12/19/2022] Open
Abstract
The delta opioid receptor (DOR), a physiologically relevant prototype for G protein-coupled receptors, is retained in intracellular compartments in neuronal cells. This retention is mediated by a nerve growth factor (NGF)-regulated checkpoint that delays the export of DOR from the trans-Golgi network. How DOR is selectively retained in the Golgi, in the midst of dynamic membrane transport and cargo export, is a fundamental unanswered question. Here we address this by investigating sequence elements on DOR that regulate DOR surface delivery, focusing on the C-terminal tail of DOR that is sufficient for NGF-mediated regulation. By systematic mutational analysis, we define conserved dual bi-arginine (RXR) motifs that are required for NGF- and phosphoinositide-regulated DOR export from intracellular compartments in neuroendocrine cells. These motifs were required to bind the coatomer protein I (COPI) complex, a vesicle coat complex that mediates primarily retrograde cargo traffic in the Golgi. Our results suggest that interactions of DOR with COPI, via atypical COPI motifs on the C-terminal tail, retain DOR in the Golgi. These interactions could provide a point of regulation of DOR export and delivery by extracellular signaling pathways.
Collapse
Affiliation(s)
- Daniel J. Shiwarski
- Department of Biological Sciences, The Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Stephanie E. Crilly
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
| | - Andrew Dates
- Department of Biological Sciences, The Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Manojkumar A. Puthenveedu
- Department of Biological Sciences, The Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
37
|
Graul M, Kisielnicka E, Rychłowski M, Verweij MC, Tobler K, Ackermann M, Wiertz EJHJ, Bieńkowska-Szewczyk K, Lipińska AD. Transmembrane regions of bovine herpesvirus 1-encoded UL49.5 and glycoprotein M regulate complex maturation and ER-Golgi trafficking. J Gen Virol 2019; 100:497-510. [PMID: 30694168 DOI: 10.1099/jgv.0.001224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bovine herpesvirus 1 (BoHV-1)-encoded UL49.5 (a homologue of herpesvirus glycoprotein N) can combine different functions, regulated by complex formation with viral glycoprotein M (gM). We aimed to identify the mechanisms governing the immunomodulatory activity of BoHV-1 UL49.5. In this study, we addressed the impact of gM/UL49.5-specific regions on heterodimer formation, folding and trafficking from the endoplasmic reticulum (ER) to the trans-Golgi network (TGN) - events previously found to be responsible for abrogation of the UL49.5-mediated inhibition of the transporter associated with antigen processing (TAP). We first established, using viral mutants, that no other viral protein could efficiently compensate for the chaperone function of UL49.5 within the complex. The cytoplasmic tail of gM, containing putative trafficking signals, was dispensable either for ER retention of gM or for the release of the complex. We constructed cell lines with stable co-expression of BoHV-1 gM with chimeric UL49.5 variants, composed of the BoHV-1 N-terminal domain fused to the transmembrane region (TM) from UL49.5 of varicella-zoster virus or TM and the cytoplasmic tail of influenza virus haemagglutinin. Those membrane-anchored N-terminal domains of UL49.5 were sufficient to form a complex, yet gM/UL49.5 folding and ER-TGN trafficking could be affected by the UL49.5 TM sequence. Finally, we found that leucine substitutions in putative glycine zipper motifs within TM helices of gM resulted in strong reduction of complex formation and decreased ability of gM to interfere with UL49.5-mediated major histocompatibility class I downregulation. These findings highlight the importance of gM/UL49.5 transmembrane domains for the biology of this conserved herpesvirus protein complex.
Collapse
Affiliation(s)
- Małgorzata Graul
- 1Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Edyta Kisielnicka
- 1Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Rychłowski
- 1Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Marieke C Verweij
- 2Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kurt Tobler
- 3Institute of Virology, University of Zurich, Zurich, Switzerland
| | | | - Emmanuel J H J Wiertz
- 4Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Krystyna Bieńkowska-Szewczyk
- 1Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Andrea D Lipińska
- 1Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
38
|
Thibodeau J, Moulefera MA, Balthazard R. On the structure–function of MHC class II molecules and how single amino acid polymorphisms could alter intracellular trafficking. Hum Immunol 2019; 80:15-31. [DOI: 10.1016/j.humimm.2018.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/01/2022]
|
39
|
Hong X, Jeyifous O, Ronilo M, Marshall J, Green WN, Standley S. A novel function for the ER retention signals in the C-terminus of kainate receptor subunit, GluK5. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:459-473. [PMID: 30339823 DOI: 10.1016/j.bbamcr.2018.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 11/30/2022]
Abstract
Classically, endoplasmic reticulum (ER) retention signals in secreted integral membrane proteins impose the requirement to assemble with other cognate subunits to form functional assemblies before they can exit the ER. We report that GluK5 has two ER retention signals in its cytoplasmic C-terminus: an arginine-based signal and a di-leucine motif previously thought to be an endocytic motif. GluK5 assembles with GluK2, but surprisingly GluK2 association does little to block the ER retention signals. We find instead that the ER retention signals are blocked by two proteins involved in intracellular trafficking, SAP97 and CASK. We show that SAP97, in the presence of CASK and the receptor complex, assumes an extended conformation. In the extended conformation, SAP97 makes its SH3 and GuK domains available to bind and sterically mask the ER retention signals in the GluK5 C-terminus. SAP97 and CASK are also necessary for sorting receptor cargoes into the local dendritic secretory pathway in neurons. We show that the ER retention signals of GluK5 play a vital role in sorting the receptor complex in the local dendritic secretory pathway in neurons. These data suggest a new role for ER retention signals in trafficking integral membrane proteins in neurons. SIGNIFICANCE: We present evidence that the ER retention signals in the kainate receptors containing GluK5 impose a requirement for sorting into local dendritic secretory pathways in neurons, as opposed to traversing the somatic Golgi apparatus. There are two ER retention signals in the C-terminus of GluK5. We show that both are blocked by physical association with SAP97 and CASK. The SH3 and GuK domains of SAP97, in the presence of CASK, bind directly to each ER retention signal and form a complex. These results support an entirely new function for ER retention signals in the C-termini of neuronal receptors, such as NMDA and kainate receptors, and define a mechanism for selective entry of receptors into local secretory pathways.
Collapse
Affiliation(s)
- Xiaoqi Hong
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91711, United States of America
| | - Okunola Jeyifous
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, United States of America
| | - Mason Ronilo
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91711, United States of America
| | - John Marshall
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, United States of America
| | - William N Green
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, United States of America
| | - Steve Standley
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91711, United States of America.
| |
Collapse
|
40
|
Vantourout P, Laing A, Woodward MJ, Zlatareva I, Apolonia L, Jones AW, Snijders AP, Malim MH, Hayday AC. Heteromeric interactions regulate butyrophilin (BTN) and BTN-like molecules governing γδ T cell biology. Proc Natl Acad Sci U S A 2018; 115:1039-1044. [PMID: 29339503 PMCID: PMC5798315 DOI: 10.1073/pnas.1701237115] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The long-held view that gamma delta (γδ) T cells in mice and humans are fundamentally dissimilar, as are γδ cells in blood and peripheral tissues, has been challenged by emerging evidence of the cells' regulation by butyrophilin (BTN) and butyrophilin-like (BTNL) molecules. Thus, murine Btnl1 and the related gene, Skint1, mediate T cell receptor (TCR)-dependent selection of murine intraepithelial γδ T cell repertoires in gut and skin, respectively; BTNL3 and BTNL8 are TCR-dependent regulators of human gut γδ cells; and BTN3A1 is essential for TCR-dependent activation of human peripheral blood Vγ9Vδ2+ T cells. However, some observations concerning BTN/Btnl molecules continue to question the extent of mechanistic conservation. In particular, murine and human gut γδ cell regulation depends on pairings of Btnl1 and Btnl6 and BTNL3 and BTNL8, respectively, whereas blood γδ cells are reported to be regulated by BTN3A1 independent of other BTNs. Addressing this paradox, we show that BTN3A2 regulates the subcellular localization of BTN3A1, including functionally important associations with the endoplasmic reticulum (ER), and is specifically required for optimal BTN3A1-mediated activation of Vγ9Vδ2+ T cells. Evidence that BTNL3/BTNL8 and Btnl1/Btnl6 likewise associate with the ER reinforces the prospect of broadly conserved mechanisms underpinning the selection and activation of γδ cells in mice and humans, and in blood and extralymphoid sites.
Collapse
Affiliation(s)
- Pierre Vantourout
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Adam Laing
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Martin J Woodward
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Iva Zlatareva
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, United Kingdom
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Luis Apolonia
- Department of Infectious Diseases, King's College London, London SE1 9RT, United Kingdom
| | - Andrew W Jones
- Mass Spectrometry Proteomics Platform, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Ambrosius P Snijders
- Mass Spectrometry Proteomics Platform, The Francis Crick Institute, London NW11AT, United Kingdom
| | - Michael H Malim
- Department of Infectious Diseases, King's College London, London SE1 9RT, United Kingdom
| | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, King's College London, London SE1 9RT, United Kingdom;
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW11AT, United Kingdom
| |
Collapse
|
41
|
Hayes M, Choudhary V, Ojha N, Shin JJ, Han GS, Carman GM, Loewen CJ, Prinz WA, Levine T. Fat storage-inducing transmembrane (FIT or FITM) proteins are related to lipid phosphatase/phosphotransferase enzymes. ACTA ACUST UNITED AC 2017; 5:88-103. [PMID: 29417057 PMCID: PMC5798408 DOI: 10.15698/mic2018.02.614] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fat storage-inducing transmembrane (FIT or FITM) proteins have been implicated in the partitioning of triacylglycerol to lipid droplets and the budding of lipid droplets from the ER. At the molecular level, the sole relevant interaction is that FITMs directly bind to triacyglycerol and diacylglycerol, but how they function at the molecular level is not known. Saccharomyces cerevisiae has two FITM homologues: Scs3p and Yft2p. Scs3p was initially identified because deletion leads to inositol auxotrophy, with an unusual sensitivity to addition of choline. This strongly suggests a role for Scs3p in phospholipid biosynthesis. Looking at the FITM family as widely as possible, we found that FITMs are widespread throughout eukaryotes, indicating presence in the last eukaryotic common ancestor. Protein alignments also showed that FITM sequences contain the active site of lipid phosphatase/phosphotransferase (LPT) enzymes. This large family transfers phosphate-containing headgroups either between lipids or in exchange for water. We confirmed the prediction that FITMs are related to LPTs by showing that single amino-acid substitutions in the presumptive catalytic site prevented their ability to rescue growth of the mutants on low inositol/high choline media when over-expressed. The substitutions also prevented rescue of other phenotypes associated with loss of FITM in yeast, including mistargeting of Opi1p, defective ER morphology, and aberrant lipid droplet budding. These results suggest that Scs3p, Yft2p and FITMs in general are LPT enzymes involved in an as yet unknown critical step in phospholipid metabolism.
Collapse
Affiliation(s)
- Matthew Hayes
- University College London Institute of Ophthalmology. 11-43 Bath Street, London, EC1V 9EL, UK
| | - Vineet Choudhary
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Namrata Ojha
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Jh Shin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gil-Soo Han
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - George M Carman
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - Christopher Jr Loewen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - William A Prinz
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy Levine
- University College London Institute of Ophthalmology. 11-43 Bath Street, London, EC1V 9EL, UK
| |
Collapse
|
42
|
Nisar SP, Lordkipanidzé M, Jones ML, Dawood BB, Murden S, Cunningham MR, Mumford AD, Wilde JT, Watson SP, Lowe GC, Mundell SJ. A novel thromboxane A2 receptor N42S variant results in reduced surface expression and platelet dysfunction. Thromb Haemost 2017; 111:923-32. [DOI: 10.1160/th13-08-0672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/19/2013] [Indexed: 11/05/2022]
Abstract
SummaryA small number of thromboxane receptor variants have been described in patients with a bleeding history that result in platelet dysfunction. We have identified a patient with a history of significant bleeding, who expresses a novel heterozygous thromboxane receptor variant that predicts an asparagine to serine substitution (N42S). This asparagine is conserved across all class A GPCRs, suggesting a vital role for receptor structure and function. We investigated the functional consequences of the TP receptor heterozygous N42S substitution by performing platelet function studies on platelet-rich plasma taken from the patient and healthy controls. We investigated the N42S mutation by expressing the wild-type (WT) and mutant receptor in human embryonic kidney (HEK) cells. Aggregation studies showed an ablation of arachidonic acid responses in the patient, whilst there was right-ward shift of the U46619 concentration response curve (CRC). Thromboxane generation was unaffected. Calcium mobilisation studies in cells lines showed a rightward shift of the U46619 CRC in N42S–expressing cells compared to WT. Radioligand binding studies revealed a reduction in BMax in platelets taken from the patient and in N42S–expressing cells, whilst cell studies confirmed poor surface expression. We have identified a novel thromboxane receptor variant, N42S, which results in platelet dysfunction due to reduced surface expression. It is associated with a significant bleeding history in the patient in whom it was identified. This is the first description of a naturally occurring variant that results in the substitution of this highly conserved residue and confirms the importance of this residue for correct GPCR function.
Collapse
|
43
|
Strategies for the etiological therapy of cystic fibrosis. Cell Death Differ 2017; 24:1825-1844. [PMID: 28937684 PMCID: PMC5635223 DOI: 10.1038/cdd.2017.126] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022] Open
Abstract
Etiological therapies aim at repairing the underlying cause of cystic fibrosis (CF), which is the functional defect of the cystic fibrosis transmembrane conductance regulator (CFTR) protein owing to mutations in the CFTR gene. Among these, the F508del CFTR mutation accounts for more than two thirds of CF cases worldwide. Two somehow antinomic schools of thought conceive CFTR repair in a different manner. According to one vision, drugs should directly target the mutated CFTR protein to increase its plasma membrane expression (correctors) or improve its ion transport function (potentiators). An alternative strategy consists in modulating the cellular environment and proteostasis networks in which the mutated CFTR protein is synthesized, traffics to its final destination, the plasma membrane, and is turned over. We will analyze distinctive advantages and drawbacks of these strategies in terms of their scientific and clinical dimensions, and we will propose a global strategy for CF research and development based on a reconciliatory approach. Moreover, we will discuss the utility of preclinical biomarkers that may guide the personalized, patient-specific implementation of CF therapies.
Collapse
|
44
|
N-Glycosylation is required for FDNC5 stabilization and irisin secretion. Biochem J 2017; 474:3167-3177. [PMID: 28733331 DOI: 10.1042/bcj20170241] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/03/2017] [Accepted: 07/20/2017] [Indexed: 12/21/2022]
Abstract
Irisin, a myokine derived from the extracellular domain of FNDC5, has been shown to mediate thermogenesis of white adipose tissue. Biochemical data have shown that N-glycosylation of FNDC5 is unlikely to affect ligand or receptor activation of irisin. The N-glycosylation of FNDC5 remains poorly understood. In the present study, we analysed N-glycosylation sites of FNDC5 and found that two potential N-glycosylation sites (Asn36 and Asn81) could indeed be occupied by N-glycan. Furthermore we showed that the lack of N-glycosylation decreases the secretion of irisin, which is relevant to the instability of FNDC5 and the deficiency of cleavage of the signal peptide. We also found that the expression level of N-glycosylated FNDC5 was elevated after myoblast differentiation. These findings show that the secretion of irisin is modulated by N-glycosylation, which in turn enhances our understanding of the secretion of glycosylated irisin.
Collapse
|
45
|
Novel mechanisms of PIEZO1 dysfunction in hereditary xerocytosis. Blood 2017; 130:1845-1856. [PMID: 28716860 DOI: 10.1182/blood-2017-05-786004] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/06/2017] [Indexed: 11/20/2022] Open
Abstract
Mutations in PIEZO1 are the primary cause of hereditary xerocytosis, a clinically heterogeneous, dominantly inherited disorder of erythrocyte dehydration. We used next-generation sequencing-based techniques to identify PIEZO1 mutations in individuals from 9 kindreds referred with suspected hereditary xerocytosis (HX) and/or undiagnosed congenital hemolytic anemia. Mutations were primarily found in the highly conserved, COOH-terminal pore-region domain. Several mutations were novel and demonstrated ethnic specificity. We characterized these mutations using genomic-, bioinformatic-, cell biology-, and physiology-based functional assays. For these studies, we created a novel, cell-based in vivo system for study of wild-type and variant PIEZO1 membrane protein expression, trafficking, and electrophysiology in a rigorous manner. Previous reports have indicated HX-associated PIEZO1 variants exhibit a partial gain-of-function phenotype with generation of mechanically activated currents that inactivate more slowly than wild type, indicating that increased cation permeability may lead to dehydration of PIEZO1-mutant HX erythrocytes. In addition to delayed channel inactivation, we found additional alterations in mutant PIEZO1 channel kinetics, differences in response to osmotic stress, and altered membrane protein trafficking, predicting variant alleles that worsen or ameliorate erythrocyte hydration. These results extend the genetic heterogeneity observed in HX and indicate that various pathophysiologic mechanisms contribute to the HX phenotype.
Collapse
|
46
|
Phillips GR, LaMassa N, Nie YM. Clustered protocadherin trafficking. Semin Cell Dev Biol 2017; 69:131-139. [PMID: 28478299 DOI: 10.1016/j.semcdb.2017.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 12/20/2022]
Abstract
The cluster of almost 60 protocadherin genes, divided into the α, β and γ subgroups, is a hallmark of vertebrate nervous system evolution. These clustered protocadherins (Pcdhs) are of interest for several reasons, one being the arrangement of the genes, which allows epigenetic regulation at the cluster and single-cell identity. Another reason is the still ambiguous effect of Pcdhs on cell-cell interaction. Unlike the case for classical cadherins, which typically mediate strong cell adhesion and formation of adherens junctions, it has been challenging to ascertain exactly how Pcdhs affect interacting cells. In some instances, Pcdhs appear to promote the association of membranes, while in other cases the Pcdhs are anti-adhesive and cause avoidance of interacting membranes. It is clear that Pcdh extracellular domains bind homophillically in an antiparallel conformation, typical of adhesive interactions. How can molecules that would seemingly bind cells together be able to promote the avoidance of membranes? It is possible that Pcdh trafficking will eventually provide insights into the role of these molecules at the cell surface. We have found that endogenous and expressed Pcdhs are generally less efficient at targeting to cell junctions and synapses than are classical cadherins. Instead, Pcdhs are prominently sequestered in the endolysosome system or other intracellular compartments. What role this trafficking plays in the unique mode of cell-cell interaction is a current topic of investigation. It is tempting to speculate that modulation of endocytosis and endolysosomal trafficking may be a part of the mechanism by which Pcdhs convert from adhesive to avoidance molecules.
Collapse
Affiliation(s)
- Greg R Phillips
- Department of Biology, Center for Developmental Neuroscience, College of Staten Island, City University of New York, United States; Program in Neuroscience, The Graduate Center, City University of New York, United States.
| | - Nicole LaMassa
- Department of Biology, Center for Developmental Neuroscience, College of Staten Island, City University of New York, United States
| | - Yan Mei Nie
- Department of Biology, Center for Developmental Neuroscience, College of Staten Island, City University of New York, United States
| |
Collapse
|
47
|
Kweon HJ, Cho JH, Jang IS, Suh BC. ASIC2a-dependent increase of ASIC3 surface expression enhances the sustained component of the currents. BMB Rep 2017; 49:542-547. [PMID: 27241858 PMCID: PMC5227295 DOI: 10.5483/bmbrep.2016.49.10.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Indexed: 12/30/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated cation channels widely expressed in the nervous system. Proton sensing by ASICs has been known to mediate pain, mechanosensation, taste transduction, learning and memory, and fear. In this study, we investigated the differential subcellular localization of ASIC2a and ASIC3 in heterologous expression systems. While ASIC2a targeted the cell surface itself, ASIC3 was mostly accumulated in the ER with partial expression in the plasma membrane. However, when ASIC3 was co-expressed with ASIC2a, its surface expression was markedly increased. By using bimolecular fluorescence complementation (BiFC) assay, we confirmed the heteromeric association between ASIC2a and ASIC3 subunits. In addition, we observed that the ASIC2a-dependent surface trafficking of ASIC3 remarkably enhanced the sustained component of the currents. Our study demonstrates that ASIC2a can increase the membrane conductance sensitivity to protons by facilitating the surface expression of ASIC3 through herteromeric assembly. [BMB Reports 2016; 49(10): 542-547].
Collapse
Affiliation(s)
- Hae-Jin Kweon
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea
| | - Jin-Hwa Cho
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Byung-Chang Suh
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea
| |
Collapse
|
48
|
Farinha CM, Canato S. From the endoplasmic reticulum to the plasma membrane: mechanisms of CFTR folding and trafficking. Cell Mol Life Sci 2017; 74:39-55. [PMID: 27699454 PMCID: PMC11107782 DOI: 10.1007/s00018-016-2387-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 01/10/2023]
Abstract
CFTR biogenesis starts with its co-translational insertion into the membrane of endoplasmic reticulum and folding of the cytosolic domains, towards the acquisition of a fully folded compact native structure. Efficiency of this process is assessed by the ER quality control system that allows the exit of folded proteins but targets unfolded/misfolded CFTR to degradation. If allowed to leave the ER, CFTR is modified at the Golgi and reaches the post-Golgi compartments to be delivered to the plasma membrane where it functions as a cAMP- and phosphorylation-regulated chloride/bicarbonate channel. CFTR residence at the membrane is a balance of membrane delivery, endocytosis, and recycling. Several adaptors, motor, and scaffold proteins contribute to the regulation of CFTR stability and are involved in continuously assessing its structure through peripheral quality control systems. Regulation of CFTR biogenesis and traffic (and its dysregulation by mutations, such as the most common F508del) determine its overall activity and thus contribute to the fine modulation of chloride secretion and hydration of epithelial surfaces. This review covers old and recent knowledge on CFTR folding and trafficking from its synthesis to the regulation of its stability at the plasma membrane and highlights how several of these steps can be modulated to promote the rescue of mutant CFTR.
Collapse
Affiliation(s)
- Carlos M Farinha
- BioISI-Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
| | - Sara Canato
- BioISI-Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| |
Collapse
|
49
|
St-Louis É, Degrandmaison J, Grastilleur S, Génier S, Blais V, Lavoie C, Parent JL, Gendron L. Involvement of the coatomer protein complex I in the intracellular traffic of the delta opioid receptor. Mol Cell Neurosci 2016; 79:53-63. [PMID: 28041939 DOI: 10.1016/j.mcn.2016.12.005] [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: 06/16/2016] [Revised: 12/01/2016] [Accepted: 12/26/2016] [Indexed: 11/16/2022] Open
Abstract
The delta opioid receptor (DOPr) is known to be mainly expressed in intracellular compartments. It remains unknown why DOPr is barely exported to the cell surface, but it seems that a substantial proportion of the immature receptor is trapped within the endoplasmic reticulum (ER) and the Golgi network. In the present study, we performed LC-MS/MS analysis to identify putative protein partners involved in the retention of DOPr. Analysis of the proteins co-immunoprecipitating with Flag-DOPr in transfected HEK293 cells revealed the presence of numerous subunits of the coatomer protein complex I (COPI), a vesicle-coating complex involved in recycling resident proteins from the Golgi back to the ER. Further analysis of the amino acid sequence of DOPr identified multiple consensus di-lysine and di-arginine motifs within the intracellular segments of DOPr. Using cell-surface ELISA and GST pulldown assays, we showed that DOPr interacts with COPI through its intracellular loops 2 and 3 (ICL2 and ICL3, respectively) and that the mutation of the K164AK166 (ICL2) or K250EK252 (ICL3) putative COPI binding sites increased the cell-surface expression of DOPr in transfected cells. Altogether, our results indicate that COPI is a binding partner of DOPr and provide a putative mechanism to explain why DOPr is highly retained inside the cells.
Collapse
Affiliation(s)
- Étienne St-Louis
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Jade Degrandmaison
- Département de médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Sébastien Grastilleur
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Samuel Génier
- Département de médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Véronique Blais
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Christine Lavoie
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Jean-Luc Parent
- Département de médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada.
| | - Louis Gendron
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Département d'anesthésiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada; Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du CHUS, Sherbrooke, Québec, Canada; Quebec Pain Research Network, Québec, Canada.
| |
Collapse
|
50
|
Acid-Sensing Ion Channel 2a (ASIC2a) Promotes Surface Trafficking of ASIC2b via Heteromeric Assembly. Sci Rep 2016; 6:30684. [PMID: 27477936 PMCID: PMC4967927 DOI: 10.1038/srep30684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/08/2016] [Indexed: 12/27/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-activated cation channels that play important roles as typical proton sensors during pathophysiological conditions and normal synaptic activities. Among the ASIC subunits, ASIC2a and ASIC2b are alternative splicing products from the same gene, ACCN1. It has been shown that ASIC2 isoforms have differential subcellular distribution: ASIC2a targets the cell surface by itself, while ASIC2b resides in the ER. However, the underlying mechanism for this differential subcellular localization remained to be further elucidated. By constructing ASIC2 chimeras, we found that the first transmembrane (TM1) domain and the proximal post-TM1 domain (17 amino acids) of ASIC2a are critical for membrane targeting of the proteins. We also observed that replacement of corresponding residues in ASIC2b by those of ASIC2a conferred proton-sensitivity as well as surface expression to ASIC2b. We finally confirmed that ASIC2b is delivered to the cell surface from the ER by forming heteromers with ASIC2a, and that the N-terminal region of ASIC2a is additionally required for the ASIC2a-dependent membrane targeting of ASIC2b. Together, our study supports an important role of ASIC2a in membrane targeting of ASIC2b.
Collapse
|