1
|
Wang Z, Liu L, Pu Y, Fang Y, Lv W, Liu W. Distinct but Redundant Roles of ER Cargo Receptors p24 and Erv29 in Facilitating Proper Secretion of Cellulases in Trichoderma reesei. Mol Microbiol 2025; 123:344-361. [PMID: 39895577 DOI: 10.1111/mmi.15343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
Trichoderma reesei represents an important industrial workhorse for (hemi)cellulase production. However, relatively little is known about the details of its secretory pathway ensuring the extremely high-level enzyme secretion and how they might be leveraged for engineering improved protein production. Here, the functions of T. reesei ER cargo receptors p24 and Erv29 in trafficking cellulase were characterised. Whereas individual deletion of p24 or erv29 resulted in only a marginal effect on extracellular cellulase secretion, distinct intracellular trafficking pathways exist for individual hydrolytic enzyme in T. reesei. Notably, the simultaneous absence of p24 and Erv29 abolished the secreted production of cellulases but not xylanases. The secretion defect was accompanied by an apparent intracellular accumulation of cellulases. Mutations of residues on the cytosolic side of p24 and Erv29 supposed to mediate COPII coat recognition also compromised cellulase secretion although the overall ER exit sites (ERES) formation did not seem to be affected. We further revealed that a VPL motif following the signal peptide of CBH2 necessitates its efficient secretion mediated by Erv29. These results indicate that two specific ER cargo receptors complement each other to mediate the proper intracellular trafficking of cellulases and thus ensuring their extracellular secretion.
Collapse
Affiliation(s)
- Zhixing Wang
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| | - Lin Liu
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| | - Yi Pu
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| | - Yu Fang
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| | - Wenhao Lv
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| | - Weifeng Liu
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, Qingdao, People's Republic of China
| |
Collapse
|
2
|
Downes KW, Zanetti G. Mechanisms of COPII coat assembly and cargo recognition in the secretory pathway. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00839-y. [PMID: 40133632 PMCID: PMC7617623 DOI: 10.1038/s41580-025-00839-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
One third of all proteins in eukaryotes transit between the endoplasmic reticulum (ER) and the Golgi to reach their functional destination inside or outside of the cell. During export, secretory proteins concentrate at transitional zones of the ER known as ER exit sites, where they are packaged into transport carriers formed by the highly conserved coat protein complex II (COPII). Despite long-standing knowledge of many of the fundamental pathways that govern traffic in the early secretory pathway, we still lack a complete mechanistic model to explain how the various steps of COPII-mediated ER exit are regulated to efficiently transport diverse cargoes. In this Review, we discuss the current understanding of the mechanisms underlying COPII-mediated vesicular transport, highlighting outstanding knowledge gaps. We focus on how coat assembly and disassembly dictate carrier morphogenesis, how COPII selectively recruits a vast number of cargo and cargo adaptors, and finally discuss how COPII mechanisms in mammals might have adapted to enable transport of large proteins.
Collapse
Affiliation(s)
- Katie W Downes
- Institute of Structural and Molecular Biology, UCL, London, UK
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK
- The Francis Crick Institute, London, UK
| | - Giulia Zanetti
- Institute of Structural and Molecular Biology, UCL, London, UK.
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
3
|
Chen X, Li F, Li X, Otto M, Chen Y, Siewers V. Model-assisted CRISPRi/a library screening reveals central carbon metabolic targets for enhanced recombinant protein production in yeast. Metab Eng 2025; 88:1-13. [PMID: 39615667 DOI: 10.1016/j.ymben.2024.11.010] [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: 04/02/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Production of recombinant proteins is regarded as an important breakthrough in the field of biomedicine and industrial biotechnology. Due to the complexity of the protein secretory pathway and its tight interaction with cellular metabolism, the application of traditional metabolic engineering tools to improve recombinant protein production faces major challenges. A systematic approach is required to generate novel design principles for superior protein secretion cell factories. Here, we applied a proteome-constrained genome-scale protein secretory model of the yeast Saccharomyces cerevisiae (pcSecYeast) to simulate α-amylase production under limited secretory capacity and predict gene targets for downregulation and upregulation to improve α-amylase production. The predicted targets were evaluated using high-throughput screening of specifically designed CRISPR interference/activation (CRISPRi/a) libraries and droplet microfluidics screening. From each library, 200 and 190 sorted clones, respectively, were manually verified. Out of them, 50% of predicted downregulation targets and 34.6% predicted upregulation targets were confirmed to improve α-amylase production. By simultaneously fine-tuning the expression of three genes in central carbon metabolism, i.e. LPD1, MDH1, and ACS1, we were able to increase the carbon flux in the fermentative pathway and α-amylase production. This study exemplifies how model-based predictions can be rapidly validated via a high-throughput screening approach. Our findings highlight novel engineering targets for cell factories and furthermore shed light on the connectivity between recombinant protein production and central carbon metabolism.
Collapse
Affiliation(s)
- Xin Chen
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark.
| | - Feiran Li
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xiaowei Li
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Maximilian Otto
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Yu Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Verena Siewers
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
4
|
Liu J, Chen H, Liu L, Meng X, Liu Q, Ye Q, Wen J, Wang T, Dong J. A cargo sorting receptor mediates chloroplast protein trafficking through the secretory pathway. THE PLANT CELL 2024; 36:3770-3786. [PMID: 38963880 PMCID: PMC11371137 DOI: 10.1093/plcell/koae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Nucleus-encoded chloroplast proteins can be transported via the secretory pathway. The molecular mechanisms underlying the trafficking of chloroplast proteins between the intracellular compartments are largely unclear, and a cargo sorting receptor has not previously been identified in the secretory pathway. Here, we report a cargo sorting receptor that is specifically present in Viridiplantae and mediates the transport of cargo proteins to the chloroplast. Using a forward genetic analysis, we identified a gene encoding a transmembrane protein (MtTP930) in barrel medic (Medicago truncatula). Mutation of MtTP930 resulted in impaired chloroplast function and a dwarf phenotype. MtTP930 is highly expressed in the aerial parts of the plant and is localized to the endoplasmic reticulum (ER) exit sites and Golgi. MtTP930 contains typical cargo sorting receptor motifs, interacts with Sar1, Sec12, and Sec24, and participates in coat protein complex II vesicular transport. Importantly, MtTP930 can recognize the cargo proteins plastidial N-glycosylated nucleotide pyrophosphatase/phosphodiesterase (MtNPP) and α-carbonic anhydrase (MtCAH) in the ER and then transport them to the chloroplast via the secretory pathway. Mutation of a homolog of MtTP930 in Arabidopsis (Arabidopsis thaliana) resulted in a similar dwarf phenotype. Furthermore, MtNPP-GFP failed to localize to chloroplasts when transgenically expressed in Attp930 protoplasts, implying that these cargo sorting receptors are conserved in plants. These findings fill a gap in our understanding of the mechanism by which chloroplast proteins are sorted and transported via the secretory pathway.
Collapse
Affiliation(s)
- Jinling Liu
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hong Chen
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Li Liu
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiangzhao Meng
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qianwen Liu
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qinyi Ye
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiangqi Wen
- Institute for Agricultural Biosciences, Oklahoma State University, Ardmore, OK 73401, USA
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Tao Wang
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiangli Dong
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
5
|
Ahmed MZ, Alqahtani AS. Cell surface expression of Ribophorin I, an endoplasmic reticulum protein, over different cell types. Int J Biol Macromol 2024; 264:130278. [PMID: 38373565 DOI: 10.1016/j.ijbiomac.2024.130278] [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: 10/01/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Ribophorin-1 serves as one of the subunits of the oligosaccharyltransferase (OST) complex located in the endoplasmic reticulum (ER). Until now, RPN-1 was considered an ER protein. However, our findings reveal that a minor fraction of RPN-1 escapes from the lumen of the ER and is ectopically expressed on the surface of different cell lines. The precise mechanism of protein translocation is unknown. The expression of RPN-1 was demonstrated through the isolation of membrane proteins using surface biotinylation and sucrose density gradient techniques. The confirmation of RPN-1 was obtained through surface staining using a specific antibody, revealing its expression on various cell lines. Additionally, we examined the expression of RPN-1 in different populations of PBMCs and observed a differential regulation of RPN-1 within PBMC subpopulations. Notably, there was a significant expression of RPN-1 on monocytes and B cells, but there was little to no population of T cells expressing RPN-1. We confirmed the expression of RPN-1 on THP-1, U937, and Jurkat cells. We also confirmed their surface expression through si-RNA knockdown. Our study shows RPN-1 expression on various cell surfaces, suggesting varied regulation among cell types. In the future, we may uncover its roles in immune function, signaling, and differentiation/proliferation.
Collapse
Affiliation(s)
- Mohammad Z Ahmed
- King Saud University College of Pharmacy, Department of Pharmacognosy, Riyadh 11451, Saudi Arabia.
| | - Ali S Alqahtani
- King Saud University College of Pharmacy, Department of Pharmacognosy, Riyadh 11451, Saudi Arabia
| |
Collapse
|
6
|
Zhang C, Yao X, Zhang Y, Zhao S, Liu J, Wu G, Yan X, Luo J. Transcriptomic Profiling Highlights the ABA Response Role of BnSIP1-1 in Brassica napus. Int J Mol Sci 2023; 24:10641. [PMID: 37445818 DOI: 10.3390/ijms241310641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
BnSIP1-1 is the first identified SIP1 (6b Interacting Protein1) subfamily gene of the trihelix transcription factor family from Brassica napus (B. napus). We previously used a reverse genetic method to reveal its abiotic stress response function in endowing plants resistance to drought and salinity, as well as ABA (Abscisic acid). However, the molecular mechanisms of BnSIP1-1 are unclear. In this study, the global transcriptome files of BnSIP1-1-overexpressing transgenic and wildtype B. napus seedlings under ABA treatment were constructed using RNA-seq. A total of 1823 and 5512 DEGs (Differentially Expressed Genes) were identified in OE vs. WT and OE_ABA vs. WT_ABA comparison groups, which included 751 and 2567 up-regulated DEGs, and 1072 and 2945 down-regulated DEGs, separately. The impact of overexpressed BnSIP1-1 on plants was amplified by ABA, indicating BnSIP1-1 was an ABA-conditioned responsive gene. More interestingly, we found the reasons for BnSIP1-1 increasing plants' insensitivity to ABA were not by regulating ABA synthesis and catabolism, but by manipulating ABA transportation, ABA signal perception and transduction, inositol phosphate metabolism, as well as endomembrane trafficking, indirectly suggesting this gene may play roles upstream of the core ABA response pathway. Our results provided new insights into improving the knowledge about the function of BnSIP1-1 and the ABA signaling mechanism in B. napus.
Collapse
Affiliation(s)
- Chi Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Xiaoqing Yao
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Yan Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Shengbo Zhao
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Jinghui Liu
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Gang Wu
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Xiaohong Yan
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| | - Junling Luo
- Key Laboratory of Biology and Genetic Improvement of Oil Crops of the Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Wuhan 430062, China
| |
Collapse
|
7
|
Fang R, Jiang Q, Jia X, Jiang Z. ARMH3-mediated recruitment of PI4KB directs Golgi-to-endosome trafficking and activation of the antiviral effector STING. Immunity 2023; 56:500-515.e6. [PMID: 36921576 DOI: 10.1016/j.immuni.2023.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/30/2022] [Accepted: 01/26/2023] [Indexed: 03/15/2023]
Abstract
The cGAS-STING pathway mediates cytoplasmic DNA-triggered innate immunity. STING activation is initiated by cyclic-GMP-AMP (cGAMP)-induced translocation from the endoplasmic reticulum and sulfated glycosaminoglycans-induced polymerization at the Golgi. Here, we examine the mechanisms underlying STING transport and activation beyond the Golgi. A genome-wide CRISPR-Cas9 screen identified Armadillo-like helical domain-containing protein 3 (ARMH3) as critical for STING activation. Upon cGAMP-triggered translocation, ARMH3 interacted with STING at the Golgi and recruited phosphatidylinositol 4-kinase beta (PI4KB) to synthesize PI4P, which directed STING Golgi-to-endosome trafficking via PI4P-binding proteins AP-1 and GGA2. Disrupting PI4P-dependent lipid transport through RNAi of other PI4P-binding proteins impaired STING activation. Consistently, disturbed lipid composition inhibited STING activation, whereas aberrantly elevated cellular PI4P led to cGAS-independent STING activation. Armh3fl/fllLyzCre/Cre mice were susceptible to DNA virus challenge in vivo. Thus, ARMH3 bridges STING and PIK4B to generate PI4P for STING transportation and activation, an interaction conserved in all eukaryotes.
Collapse
Affiliation(s)
- Run Fang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qifei Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xinying Jia
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
8
|
Targeted Protein Unfolding at the Golgi Apparatus. Methods Mol Biol 2022; 2557:645-659. [PMID: 36512243 DOI: 10.1007/978-1-0716-2639-9_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Maintaining protein homeostasis (proteostasis) is vital to cellular and organismal health. How the Golgi apparatus, the central protein maturation and sorting station in the cell, manages misfolded proteins to maintain proteostasis is still poorly understood. Here we present a strategy for targeted protein unfolding at the Golgi that enables studying Golgi-related protein quality control and stress-signaling pathways. Targeted protein unfolding is induced by small molecule-based chemical biology approaches-hydrophobic tagging and the use of a destabilization domain. Imaging studies allow visualizing quality control (QC) phenotypes, such as the formation of QC carriers and Golgi-to-endoplasmic reticulum trafficking, and correlating these phenotypes with other trafficking processes.
Collapse
|
9
|
Gomez-Navarro N, Maldutyte J, Poljak K, Peak-Chew SY, Orme J, Bisnett BJ, Lamb CH, Boyce M, Gianni D, Miller EA. Selective inhibition of protein secretion by abrogating receptor-coat interactions during ER export. Proc Natl Acad Sci U S A 2022; 119:e2202080119. [PMID: 35901214 PMCID: PMC9351455 DOI: 10.1073/pnas.2202080119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 01/03/2023] Open
Abstract
Protein secretion is an essential process that drives cell growth, movement, and communication. Protein traffic within the secretory pathway occurs via transport intermediates that bud from one compartment and fuse with a downstream compartment to deliver their contents. Here, we explore the possibility that protein secretion can be selectively inhibited by perturbing protein-protein interactions that drive capture into transport vesicles. Human proprotein convertase subtilisin/kexin type 9 (PCSK9) is a determinant of cholesterol metabolism whose secretion is mediated by a specific cargo adaptor protein, SEC24A. We map a series of protein-protein interactions between PCSK9, its endoplasmic reticulum (ER) export receptor SURF4, and SEC24A that mediate secretion of PCSK9. We show that the interaction between SURF4 and SEC24A can be inhibited by 4-phenylbutyrate (4-PBA), a small molecule that occludes a cargo-binding domain of SEC24. This inhibition reduces secretion of PCSK9 and additional SURF4 clients that we identify by mass spectrometry, leaving other secreted cargoes unaffected. We propose that selective small-molecule inhibition of cargo recognition by SEC24 is a potential therapeutic intervention for atherosclerosis and other diseases that are modulated by secreted proteins.
Collapse
Affiliation(s)
- Natalia Gomez-Navarro
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Julija Maldutyte
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Kristina Poljak
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Sew-Yeu Peak-Chew
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Jonathon Orme
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Brittany J. Bisnett
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Caitlin H. Lamb
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Davide Gianni
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Elizabeth A. Miller
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| |
Collapse
|
10
|
Bean BDM, Mulvihill CJ, Garge RK, Boutz DR, Rousseau O, Floyd BM, Cheney W, Gardner EC, Ellington AD, Marcotte EM, Gollihar JD, Whiteway M, Martin VJJ. Functional expression of opioid receptors and other human GPCRs in yeast engineered to produce human sterols. Nat Commun 2022; 13:2882. [PMID: 35610225 PMCID: PMC9130329 DOI: 10.1038/s41467-022-30570-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
The yeast Saccharomyces cerevisiae is powerful for studying human G protein-coupled receptors as they can be coupled to its mating pathway. However, some receptors, including the mu opioid receptor, are non-functional, which may be due to the presence of the fungal sterol ergosterol instead of cholesterol. Here we engineer yeast to produce cholesterol and introduce diverse mu, delta, and kappa opioid receptors to create sensitive opioid biosensors that recapitulate agonist binding profiles and antagonist inhibition. Additionally, human mu opioid receptor variants, including those with clinical relevance, largely display expected phenotypes. By testing mu opioid receptor-based biosensors with systematically adjusted cholesterol biosynthetic intermediates, we relate sterol profiles to biosensor sensitivity. Finally, we apply sterol-modified backgrounds to other human receptors revealing sterol influence in SSTR5, 5-HTR4, FPR1, and NPY1R signaling. This work provides a platform for generating human G protein-coupled receptor-based biosensors, facilitating receptor deorphanization and high-throughput screening of receptors and effectors.
Collapse
Affiliation(s)
- Björn D M Bean
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Colleen J Mulvihill
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Riddhiman K Garge
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Daniel R Boutz
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
- DEVCOM Army Research Laboratory-South, Austin, 78712, TX, USA
| | - Olivier Rousseau
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Brendan M Floyd
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - William Cheney
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Elizabeth C Gardner
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew D Ellington
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Edward M Marcotte
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jimmy D Gollihar
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
- DEVCOM Army Research Laboratory-South, Austin, 78712, TX, USA.
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| | - Malcolm Whiteway
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Vincent J J Martin
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada.
| |
Collapse
|
11
|
Manzano-Lopez J, Rodriguez-Gallardo S, Sabido-Bozo S, Cortes-Gomez A, Perez-Linero AM, Lucena R, Cordones-Romero A, Lopez S, Aguilera-Romero A, Muñiz M. Crosslinking assay to study a specific cargo-coat interaction through a transmembrane receptor in the secretory pathway. PLoS One 2022; 17:e0263617. [PMID: 35143573 PMCID: PMC8830656 DOI: 10.1371/journal.pone.0263617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/21/2022] [Indexed: 12/03/2022] Open
Abstract
Intracellular trafficking through the secretory organelles depends on transient interactions between cargo proteins and transport machinery. Cytosolic coat protein complexes capture specific luminal cargo proteins for incorporation into transport vesicles by interacting with them indirectly through a transmembrane adaptor or cargo receptor. Due to their transient nature, it is difficult to study these specific ternary protein interactions just using conventional native co-immunoprecipitation. To overcome this technical challenge, we have applied a crosslinking assay to stabilize the transient and/or weak protein interactions. Here, we describe a protocol of protein crosslinking and co-immunoprecipitation, which was employed to prove the indirect interaction in the endoplasmic reticulum of a luminal secretory protein with a selective subunit of the cytosolic COPII coat through a specific transmembrane cargo receptor. This method can be extended to address other transient ternary interactions between cytosolic proteins and luminal or extracellular proteins through a transmembrane receptor within the endomembrane system.
Collapse
Affiliation(s)
- Javier Manzano-Lopez
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- * E-mail: (JM-L); (AA-R); (MM)
| | - Sofia Rodriguez-Gallardo
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Susana Sabido-Bozo
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Alejandro Cortes-Gomez
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Ana Maria Perez-Linero
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rafael Lucena
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio Cordones-Romero
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Sergio Lopez
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Auxiliadora Aguilera-Romero
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- * E-mail: (JM-L); (AA-R); (MM)
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- * E-mail: (JM-L); (AA-R); (MM)
| |
Collapse
|
12
|
Zhang N, Zabotina OA. Critical Determinants in ER-Golgi Trafficking of Enzymes Involved in Glycosylation. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11030428. [PMID: 35161411 PMCID: PMC8840164 DOI: 10.3390/plants11030428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 05/03/2023]
Abstract
All living cells generate structurally complex and compositionally diverse spectra of glycans and glycoconjugates, critical for organismal evolution, development, functioning, defense, and survival. Glycosyltransferases (GTs) catalyze the glycosylation reaction between activated sugar and acceptor substrate to synthesize a wide variety of glycans. GTs are distributed among more than 130 gene families and are involved in metabolic processes, signal pathways, cell wall polysaccharide biosynthesis, cell development, and growth. Glycosylation mainly takes place in the endoplasmic reticulum (ER) and Golgi, where GTs and glycosidases involved in this process are distributed to different locations of these compartments and sequentially add or cleave various sugars to synthesize the final products of glycosylation. Therefore, delivery of these enzymes to the proper locations, the glycosylation sites, in the cell is essential and involves numerous secretory pathway components. This review presents the current state of knowledge about the mechanisms of protein trafficking between ER and Golgi. It describes what is known about the primary components of protein sorting machinery and trafficking, which are recognition sites on the proteins that are important for their interaction with the critical components of this machinery.
Collapse
|
13
|
Zhao GD, Gao R, Hou XT, Zhang H, Chen XT, Luo JQ, Yang HF, Chen T, Shen X, Yang SC, Wu CL, Huang G. Endoplasmic Reticulum Stress Mediates Renal Tubular Vacuolation in BK Polyomavirus-Associated Nephropathy. Front Endocrinol (Lausanne) 2022; 13:834187. [PMID: 35464062 PMCID: PMC9027570 DOI: 10.3389/fendo.2022.834187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/04/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE This study aimed to explore the molecular mechanism of cytoplasmic vacuolation caused by BK polyomavirus (BKPyV) and thus search for potential target for drug repurposing. METHODS Morphological features of BK polyomavirus-associated nephropathy (BKPyVAN) were studied under light and electron microscopes. Microarray datasets GSE75693, GSE47199, and GSE72925 were integrated by ComBat, and differentially expressed genes (DEGs) were analyzed using limma. Furthermore, the endoplasmic reticulum (ER)-related genes obtained from GenCLiP 2.0 were intersected with DEGs. GO and KEGG enrichment pathways were performed with intersection genes by R package clusterProfiler. The single-cell RNA sequencing (scRNA-seq) from a BKPyVAN recipient was analyzed with a dataset (GSE140989) downloaded from Gene Expression Omnibus (GEO) as control for gene set variation analysis (GSVA). Immunohistochemistry and electron microscopy of kidney sections from drug-induced ERS mouse models were performed to explore the association of ERS and renal tubular vacuolation. Protein-protein interaction (PPI) network of the intersection genes was constructed to identify hub target. AutoDock was used to screen Food and Drug Administration (FDA)-approved drugs that potentially targeted hub gene. RESULTS Light and electron microscopes exhibited obvious intranuclear inclusions, vacuoles, and virus particles in BKPyV-infected renal tubular cells. Transcriptome analysis revealed 629 DEGs between samples of BKPyVAN and stable transplanted kidneys, of which 16 were ER-associated genes. GO analysis with the intersection genes illustrated that ERS-related pathways were significantly involved, and KEGG analysis showed a prominent enrichment of MAPK, Toll-like receptor, and chemokine signaling pathways. GSVA analysis of the proximal tubule revealed similar pathways enrichment. An electron microscope image of the kidney from ERS mouse models showed an obvious renal tubular vacuolation with prominent activation of ERS markers verified by immunohistochemistry. Furthermore, DDIT3 was identified as the hub gene based on PPI analysis, and ZINCOOOOO1531009 (Risedronate) was indicated to be a potential drug for DDIT3. CONCLUSION ERS was involved in renal tubular cytoplasmic vacuolation in BKPyVAN recipients. Risedronate was screened as a potential drug for BKPyVAN by targeting DDIT3.
Collapse
Affiliation(s)
- Guo-Dong Zhao
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rong Gao
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiao-Tao Hou
- Department of Renal Pathology, King Medical Diagnostics Center, Guangzhou, China
| | - Hui Zhang
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xu-Tao Chen
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jin-Quan Luo
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui-Fei Yang
- Department of Pathology, Fuda Cancer Hospital·Jinan University, Guangzhou, China
| | - Tong Chen
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xue Shen
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shi-Cong Yang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Cheng-Lin Wu
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Gang Huang, ; Cheng-Lin Wu,
| | - Gang Huang
- Department of Organ Transplant, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Gang Huang, ; Cheng-Lin Wu,
| |
Collapse
|
14
|
Fenech EJ, Ben-Dor S, Schuldiner M. Double the Fun, Double the Trouble: Paralogs and Homologs Functioning in the Endoplasmic Reticulum. Annu Rev Biochem 2021; 89:637-666. [PMID: 32569522 DOI: 10.1146/annurev-biochem-011520-104831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The evolution of eukaryotic genomes has been propelled by a series of gene duplication events, leading to an expansion in new functions and pathways. While duplicate genes may retain some functional redundancy, it is clear that to survive selection they cannot simply serve as a backup but rather must acquire distinct functions required for cellular processes to work accurately and efficiently. Understanding these differences and characterizing gene-specific functions is complex. Here we explore different gene pairs and families within the context of the endoplasmic reticulum (ER), the main cellular hub of lipid biosynthesis and the entry site for the secretory pathway. Focusing on each of the ER functions, we highlight specificities of related proteins and the capabilities conferred to cells through their conservation. More generally, these examples suggest why related genes have been maintained by evolutionary forces and provide a conceptual framework to experimentally determine why they have survived selection.
Collapse
Affiliation(s)
- Emma J Fenech
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Shifra Ben-Dor
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel;
| |
Collapse
|
15
|
Yoshioka H, Okita S, Nakano M, Minamizaki T, Nubukiyo A, Sotomaru Y, Bonnelye E, Kozai K, Tanimoto K, Aubin JE, Yoshiko Y. Single-Cell RNA-Sequencing Reveals the Breadth of Osteoblast Heterogeneity. JBMR Plus 2021; 5:e10496. [PMID: 34189385 PMCID: PMC8216137 DOI: 10.1002/jbm4.10496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
The current paradigm of osteoblast fate is that the majority undergo apoptosis, while some further differentiate into osteocytes and others flatten and cover bone surfaces as bone lining cells. Osteoblasts have been described to exhibit heterogeneous expression of a variety of osteoblast markers at both transcriptional and protein levels. To explore further this heterogeneity and its biological significance, Venus‐positive (Venus+) cells expressing the fluorescent protein Venus under the control of the 2.3‐kb Col1a1 promoter were isolated from newborn mouse calvariae and subjected to single‐cell RNA sequencing. Functional annotation of the genes expressed in 272 Venus+ single cells indicated that Venus+ cells are osteoblasts that can be categorized into four clusters. Of these, three clusters (clusters 1 to 3) exhibited similarities in their expression of osteoblast markers, while one (cluster 4) was distinctly different. We identified a total of 1920 cluster‐specific genes and pseudotime ordering analyses based on established concepts and known markers showed that clusters 1 to 3 captured osteoblasts at different maturational stages. Analysis of gene co‐expression networks showed that genes involved in protein synthesis and protein trafficking between endoplasmic reticulum (ER) and Golgi are active in these clusters. However, the cells in these clusters were also defined by extensive heterogeneity of gene expression, independently of maturational stage. Cells of cluster 4 expressed Cd34 and Cxcl12 with relatively lower levels of osteoblast markers, suggesting that this cell type differs from actively bone‐forming osteoblasts and retain or reacquire progenitor properties. Based on expression and machine learning analyses of the transcriptomes of individual osteoblasts, we also identified genes that may be useful as new markers of osteoblast maturational stages. Taken together, our data show much more extensive heterogeneity of osteoblasts than previously documented, with gene profiles supporting diversity of osteoblast functional activities and developmental fates. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hirotaka Yoshioka
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan.,Department of Anatomy School of Medicine, International University of Health and Welfare Chiba Japan
| | - Saki Okita
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan.,Department of Craniofacial and Developmental Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Masashi Nakano
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan.,Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan.,Department of Pediatric Dentistry Hiroshima University Hospital Hiroshima Japan
| | - Tomoko Minamizaki
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Asako Nubukiyo
- Natural Science Center of Basic Research and Development Hiroshima University Hiroshima Japan
| | - Yusuke Sotomaru
- Natural Science Center of Basic Research and Development Hiroshima University Hiroshima Japan
| | - Edith Bonnelye
- CNRS ERL 6001/INSERM U1232 Institut de Cancérologie de l'Ouest Saint-Herblain France
| | - Katsuyuki Kozai
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Kotaro Tanimoto
- Department of Craniofacial and Developmental Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Jane E Aubin
- Department of Molecular Genetics University of Toronto Toronto Canada
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| |
Collapse
|
16
|
Gomez-Navarro N, Melero A, Li XH, Boulanger J, Kukulski W, Miller EA. Cargo crowding contributes to sorting stringency in COPII vesicles. J Cell Biol 2021; 219:151777. [PMID: 32406500 PMCID: PMC7300426 DOI: 10.1083/jcb.201806038] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 03/11/2020] [Accepted: 04/24/2020] [Indexed: 02/05/2023] Open
Abstract
Accurate maintenance of organelle identity in the secretory pathway relies on retention and retrieval of resident proteins. In the endoplasmic reticulum (ER), secretory proteins are packaged into COPII vesicles that largely exclude ER residents and misfolded proteins by mechanisms that remain unresolved. Here we combined biochemistry and genetics with correlative light and electron microscopy (CLEM) to explore how selectivity is achieved. Our data suggest that vesicle occupancy contributes to ER retention: in the absence of abundant cargo, nonspecific bulk flow increases. We demonstrate that ER leakage is influenced by vesicle size and cargo occupancy: overexpressing an inert cargo protein or reducing vesicle size restores sorting stringency. We propose that cargo recruitment into vesicles creates a crowded lumen that drives selectivity. Retention of ER residents thus derives in part from the biophysical process of cargo enrichment into a constrained spherical membrane-bound carrier.
Collapse
Affiliation(s)
| | - Alejandro Melero
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Xiao-Han Li
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Jérôme Boulanger
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Wanda Kukulski
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
17
|
Alva TR, Riera M, Chartron JW. Translational landscape and protein biogenesis demands of the early secretory pathway in Komagataella phaffii. Microb Cell Fact 2021; 20:19. [PMID: 33472617 PMCID: PMC7816318 DOI: 10.1186/s12934-020-01489-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/29/2020] [Indexed: 11/24/2022] Open
Abstract
Background Eukaryotes use distinct networks of biogenesis factors to synthesize, fold, monitor, traffic, and secrete proteins. During heterologous expression, saturation of any of these networks may bottleneck titer and yield. To understand the flux through various routes into the early secretory pathway, we quantified the global and membrane-associated translatomes of Komagataella phaffii. Results By coupling Ribo-seq with long-read mRNA sequencing, we generated a new annotation of protein-encoding genes. By using Ribo-seq with subcellular fractionation, we quantified demands on co- and posttranslational translocation pathways. During exponential growth in rich media, protein components of the cell-wall represent the greatest number of nascent chains entering the ER. Transcripts encoding the transmembrane protein PMA1 sequester more ribosomes at the ER membrane than any others. Comparison to Saccharomyces cerevisiae reveals conservation in the resources allocated by gene ontology, but variation in the diversity of gene products entering the secretory pathway. Conclusion A subset of host proteins, particularly cell-wall components, impose the greatest biosynthetic demands in the early secretory pathway. These proteins are potential targets in strain engineering aimed at alleviating bottlenecks during heterologous protein production.
Collapse
Affiliation(s)
- Troy R Alva
- Department of Bioengineering, University of California, Riverside, 92521, United States of America.
| | - Melanie Riera
- Department of Bioengineering, University of California, Riverside, 92521, United States of America
| | - Justin W Chartron
- Department of Bioengineering, University of California, Riverside, 92521, United States of America.,Protabit LLC, 1010 E Union St Suite 110, Pasadena, California, 91106, United States of America
| |
Collapse
|
18
|
Chang YJ, Chen KW, Chen L. Mitochondrial ROS1 Increases Mitochondrial Fission and Respiration in Oral Squamous Cancer Carcinoma. Cancers (Basel) 2020; 12:cancers12102845. [PMID: 33019722 PMCID: PMC7599653 DOI: 10.3390/cancers12102845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The clinical efficacy of anti-epidermal growth factor receptor (EGFR) antibody cetuximab for oral squamous cell carcinomas (OSCCs) is low. We previously reported that an increased oncogenic ROS proto-oncogene 1 (ROS1) is responsible for the invasiveness and metastasis of OSCC. This study demonstrates for the first time that ROS1, a receptor tyrosine kinase, can localize to mitochondria. Mitochondrial ROS1 in the highly invasive OSCC promotes mitochondrial fission, enhances mitochondrial oxidative phosphorylation and ATP production but reduces mitochondrial biogenesis. These findings highlight the novel function of ROS1 in mitochondrial morphogenesis and metabolic adaptation to promote OSCC invasiveness. Abstract Increased ROS proto-oncogene 1 (ROS1) expression has been implicated in the invasiveness of human oral squamous cell carcinoma (OSCC). The cellular distribution of ROS1 has long-been assumed at the plasma membrane. However, a previous work reported a differential cellular distribution of mutant ROS1 derived from chromosomal translocation, resulting in increased carcinogenesis. We thus hypothesized that cellular distribution of upregulated ROS1 in OSCC may correlate with invasiveness. We found that ROS1 can localize to mitochondria in the highly invasive OSCC and identified a mitochondria-targeting signal sequence in ROS1. We also demonstrated that ROS1 targeting to mitochondria is required for mitochondrial fission phenotype in the highly invasive OSCC cells. OSCC cells expressing high levels of ROS1 consumed more oxygen and had increased levels of cellular ATP levels. Our results also revealed that ROS1 regulates mitochondrial biogenesis and cellular metabolic plasticity. Together, these findings demonstrate that ROS1 targeting to mitochondria enhances OSCC invasion through regulating mitochondrial morphogenesis and cellular respiratory.
Collapse
Affiliation(s)
- Yu-Jung Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Correspondence: (Y.-J.C.); (L.C.); Tel.: +886-3-571-5131#33473 (Y.-J.C.); +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| | - Kuan-Wei Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (Y.-J.C.); (L.C.); Tel.: +886-3-571-5131#33473 (Y.-J.C.); +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| |
Collapse
|
19
|
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
|
20
|
Sun X, Tie HC, Chen B, Lu L. Glycans function as a Golgi export signal to promote the constitutive exocytic trafficking. J Biol Chem 2020; 295:14750-14762. [PMID: 32826314 PMCID: PMC7586228 DOI: 10.1074/jbc.ra120.014476] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/17/2020] [Indexed: 11/15/2022] Open
Abstract
Most proteins in the secretory pathway are glycosylated. However, the role of glycans in membrane trafficking is still unclear. Here, we discovered that transmembrane secretory cargos, such as interleukin 2 receptor α subunit or Tac, transferrin receptor, and cluster of differentiation 8a, unexpectedly displayed substantial Golgi localization when their O-glycosylation was compromised. By quantitatively measuring their Golgi residence times, we found that the observed Golgi localization of O-glycan–deficient cargos is due to their slow Golgi export. Using a superresolution microscopy method that we previously developed, we revealed that O-glycan–deficient Tac chimeras localize at the interior of the trans-Golgi cisternae. O-Glycans were observed to be both necessary and sufficient for the efficient Golgi export of Tac chimeras. By sequentially introducing O-glycosylation sites to ST6GAL1, we demonstrated that O-glycan's effect on Golgi export is probably additive. Finally, the finding that N-glycosylated GFP substantially reduces the Golgi residence time of a Tac chimera suggests that N-glycans might have a similar effect. Therefore, both O- and N-glycans might function as a generic Golgi export signal at the trans-Golgi to promote the constitutive exocytic trafficking.
Collapse
Affiliation(s)
- Xiuping Sun
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Hieng Chiong Tie
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Bing Chen
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Lei Lu
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
21
|
Hepatitis B Virus Exploits ERGIC-53 in Conjunction with COPII to Exit Cells. Cells 2020; 9:cells9081889. [PMID: 32806600 PMCID: PMC7464876 DOI: 10.3390/cells9081889] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
Several decades after its discovery, the hepatitis B virus (HBV) still displays one of the most successful pathogens in human populations worldwide. The identification and characterization of interactions between cellular and pathogenic components are essential for the development of antiviral treatments. Due to its small-sized genome, HBV highly depends on cellular functions to produce and export progeny particles. Deploying biochemical-silencing methods and molecular interaction studies in HBV-expressing liver cells, we herein identified the cellular ERGIC-53, a high-mannose-specific lectin, and distinct components of the endoplasmic reticulum (ER) export machinery COPII as crucial factors of viral trafficking and egress. Whereas the COPII subunits Sec24A, Sec23B and Sar1 are needed for both viral and subviral HBV particle exit, ERGIC-53 appears as an exclusive element of viral particle propagation, therefore interacting with the N146-glycan of the HBV envelope in a productive manner. Cell-imaging studies pointed to ER-derived, subcellular compartments where HBV assembly initiates. Moreover, our findings provide evidence that HBV exploits the functions of ERGIC-53 and Sec24A after the envelopment of nucleocapsids at these compartments in conjunction with endosomal sorting complexes required for transport (ESCRT) components. These data reveal novel insights into HBV assembly and trafficking, illustrating therapeutic prospects for intervening with the viral life cycle.
Collapse
|
22
|
Lopez S, Perez-Linero AM, Manzano-Lopez J, Sabido-Bozo S, Cortes-Gomez A, Rodriguez-Gallardo S, Aguilera-Romero A, Goder V, Muñiz M. Dual Independent Roles of the p24 Complex in Selectivity of Secretory Cargo Export from the Endoplasmic Reticulum. Cells 2020; 9:cells9051295. [PMID: 32456004 PMCID: PMC7291304 DOI: 10.3390/cells9051295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 11/22/2022] Open
Abstract
The cellular mechanisms that ensure the selectivity and fidelity of secretory cargo protein transport from the endoplasmic reticulum (ER) to the Golgi are still not well understood. The p24 protein complex acts as a specific cargo receptor for GPI-anchored proteins by facilitating their ER exit through a specialized export pathway in yeast. In parallel, the p24 complex can also exit the ER using the general pathway that exports the rest of secretory proteins with their respective cargo receptors. Here, we show biochemically that the p24 complex associates at the ER with other cargo receptors in a COPII-dependent manner, forming high-molecular weight multireceptor complexes. Furthermore, live cell imaging analysis reveals that the p24 complex is required to retain in the ER secretory cargos when their specific receptors are absent. This requirement does not involve neither the unfolded protein response nor the retrograde transport from the Golgi. Our results suggest that, in addition to its role as a cargo receptor in the specialized GPI-anchored protein pathway, the p24 complex also plays an independent role in secretory cargo selectivity during its exit through the general ER export pathway, preventing the non-selective bulk flow of native secretory cargos. This mechanism would ensure receptor-regulated cargo transport, providing an additional layer of regulation of secretory cargo selectivity during ER export.
Collapse
Affiliation(s)
- Sergio Lopez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Ana Maria Perez-Linero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
| | - Javier Manzano-Lopez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
| | - Susana Sabido-Bozo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Alejandro Cortes-Gomez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Sofia Rodriguez-Gallardo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Auxiliadora Aguilera-Romero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Veit Goder
- Department of Genetics, University of Seville, 41012 Seville, Spain;
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
- Correspondence: ; Tel.: +34-954556529
| |
Collapse
|
23
|
Small Molecule Targets TMED9 and Promotes Lysosomal Degradation to Reverse Proteinopathy. Cell 2020; 178:521-535.e23. [PMID: 31348885 DOI: 10.1016/j.cell.2019.07.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 04/19/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.
Collapse
|
24
|
Otamendi A, Perez-de-Nanclares-Arregi E, Oiartzabal-Arano E, Cortese MS, Espeso EA, Etxebeste O. Developmental regulators FlbE/D orchestrate the polarity site-to-nucleus dynamics of the fungal bZIP transcription factor FlbB. Cell Mol Life Sci 2019; 76:4369-4390. [PMID: 31065746 PMCID: PMC11105705 DOI: 10.1007/s00018-019-03121-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
Abstract
Permanently polarized cells have developed transduction mechanisms linking polarity sites with gene regulation in the nucleus. In neurons, one mechanism is based on long-distance retrograde migration of transcription factors (TFs). Aspergillus nidulans FlbB is the only known fungal TF shown to migrate retrogradely to nuclei from the polarized region of fungal cells known as hyphae. There, FlbB controls developmental transitions by triggering the production of asexual multicellular structures. FlbB dynamics in hyphae is orchestrated by regulators FlbE and FlbD. At least three FlbE domains are involved in the acropetal transport of FlbB, with a final MyoE/actin filament-dependent step from the subapex to the apex. Experiments employing a T2A viral peptide-containing chimera (FlbE::mRFP::T2A::FlbB::GFP) suggest that apical FlbB/FlbE interaction is inhibited to initiate a dynein-dependent FlbB transport to nuclei. FlbD controls the nuclear accumulation of FlbB through a cMyb domain and a C-terminal LxxLL motif. Overall, results elucidate a highly dynamic pattern of FlbB interactions, which enable timely developmental induction. Furthermore, this system establishes a reference for TF-based long-distance signaling in permanently polarized cells.
Collapse
Affiliation(s)
- Ainara Otamendi
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Elixabet Perez-de-Nanclares-Arregi
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Elixabet Oiartzabal-Arano
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Marc S Cortese
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Eduardo A Espeso
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Oier Etxebeste
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain.
| |
Collapse
|
25
|
Affiliation(s)
- J Paul Luzio
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
26
|
Perrier A, Bonnin A, Desmarets L, Danneels A, Goffard A, Rouillé Y, Dubuisson J, Belouzard S. The C-terminal domain of the MERS coronavirus M protein contains a trans-Golgi network localization signal. J Biol Chem 2019; 294:14406-14421. [PMID: 31399512 PMCID: PMC6768645 DOI: 10.1074/jbc.ra119.008964] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/06/2019] [Indexed: 11/23/2022] Open
Abstract
Coronavirus M proteins represent the major protein component of the viral
envelope. They play an essential role during viral assembly by interacting with
all of the other structural proteins. Coronaviruses bud into the endoplasmic
reticulum (ER)–Golgi intermediate compartment (ERGIC), but the mechanisms
by which M proteins are transported from their site of synthesis, the ER, to the
budding site remain poorly understood. Here, we investigated the intracellular
trafficking of the Middle East respiratory syndrome coronavirus (MERS-CoV) M
protein. Subcellular localization analyses revealed that the MERS-CoV M protein
is retained intracellularly in the trans-Golgi network (TGN),
and we identified two motifs in the distal part of the C-terminal domain as
being important for this specific localization. We identified the first motif as
a functional diacidic DxE ER export signal, because substituting Asp-211 and
Glu-213 with alanine induced retention of the MERS-CoV M in the ER. The second
motif, 199KxGxYR204, was responsible for retaining the M
protein in the TGN. Substitution of this motif resulted in MERS-CoV M leakage
toward the plasma membrane. We further confirmed the role of
199KxGxYR204 as a TGN retention signal by using
chimeras between MERS-CoV M and the M protein of infectious bronchitis virus
(IBV). Our results indicated that the C-terminal domains of both proteins
determine their specific localization, namely TGN and
ERGIC/cis-Golgi for MERS-M and IBV-M, respectively. Our
findings indicate that MERS-CoV M protein localizes to the TGN because of the
combined presence of an ER export signal and a TGN retention motif.
Collapse
Affiliation(s)
- Anabelle Perrier
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Ariane Bonnin
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Lowiese Desmarets
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Adeline Danneels
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Anne Goffard
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Yves Rouillé
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Jean Dubuisson
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Sandrine Belouzard
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
27
|
Hellerschmied D, Serebrenik YV, Shao L, Burslem GM, Crews CM. Protein folding state-dependent sorting at the Golgi apparatus. Mol Biol Cell 2019; 30:2296-2308. [PMID: 31166830 PMCID: PMC6743468 DOI: 10.1091/mbc.e19-01-0069] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
In eukaryotic cells, organelle-specific protein quality control (PQC) is critical for maintaining cellular homeostasis. Despite the Golgi apparatus being the major protein processing and sorting site within the secretory pathway, how it contributes to PQC has remained largely unknown. Using different chemical biology-based protein unfolding systems, we reveal the segregation of unfolded proteins from folded proteins in the Golgi. Quality control (QC) substrates are subsequently exported in distinct carriers, which likely contain unfolded proteins as well as highly oligomerized cargo that mimic protein aggregates. At an additional sorting step, oligomerized proteins are committed to lysosomal degradation, while unfolded proteins localize to the endoplasmic reticulum (ER) and associate with chaperones. These results highlight the existence of checkpoints at which QC substrates are selected for Golgi export and lysosomal degradation. Our data also suggest that the steady-state ER localization of misfolded proteins, observed for several disease-causing mutants, may have different origins.
Collapse
Affiliation(s)
| | | | - Lin Shao
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520
| | | | - Craig M. Crews
- Department of Molecular, Cellular and Developmental Biology
- Department of Chemistry, Yale University, New Haven, CT 06511
- Department of Pharmacology, Yale University, New Haven, CT 06511
| |
Collapse
|
28
|
Yoo W, Cho EB, Kim S, Yoon JB. The E3 ubiquitin ligase MARCH2 regulates ERGIC3-dependent trafficking of secretory proteins. J Biol Chem 2019; 294:10900-10912. [PMID: 31142615 DOI: 10.1074/jbc.ra119.007435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/17/2019] [Indexed: 11/06/2022] Open
Abstract
The E3 ubiquitin ligase membrane-associated ring-CH-type finger 2 (MARCH2) is known to be involved in intracellular vesicular trafficking, but its role in the early secretory pathway between the endoplasmic reticulum (ER) and Golgi compartments is largely unknown. Human ER-Golgi intermediate compartment protein 2 (ERGIC2) and ERGIC3 are orthologs of Erv41 and Erv46 in yeast, proteins that form a heteromeric complex, cycle between the ER and Golgi, and function as cargo receptors in both anterograde and retrograde protein trafficking. Here, we report that MARCH2 directs ubiquitination and subsequent degradation of ERGIC3 and that MARCH2 depletion increases endogenous ERGIC3 levels. We provide evidence that the lysine residues at positions 6 and 8 of ERGIC3 are the major sites of MARCH2-mediated ubiquitination. Of note, MARCH2 did not significantly decrease the levels of an ERGIC3 variant with lysine-to-arginine substitutions at residues 6 and 8. We also show that ERGIC3 binds to itself or to ERGIC2, whereas ERGIC2 is unable to interact with itself. Our results indicate that α1-antitrypsin and haptoglobin are likely to be cargo proteins of ERGIC3. We further observed that α1-antitrypsin and haptoglobin specifically bind to ERGIC3 and that ERGIC3 depletion decreases their secretion. Moreover, MARCH2 reduced secretion of α1-antitrypsin and haptoglobin, and coexpression of the ubiquitination-resistant ERGIC3 variant largely restored their secretion, suggesting that MARCH2-mediated ERGIC3 ubiquitination is the major cause of the decrease in trafficking of ERGIC3-binding secretory proteins. Our findings provide detailed insights into the regulation of the early secretory pathway by MARCH2 and into ERGIC3 function.
Collapse
Affiliation(s)
- Wonjin Yoo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea and
| | - Eun-Bee Cho
- Department of Medical Lifesciences, Catholic University of Korea, Seoul 06591, Korea
| | - Sungjoo Kim
- Department of Medical Lifesciences, Catholic University of Korea, Seoul 06591, Korea
| | - Jong-Bok Yoon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea and.
| |
Collapse
|
29
|
Disruption of Structural Disulfides of Coagulation FXIII-B Subunit; Functional Implications for a Rare Bleeding Disorder. Int J Mol Sci 2019; 20:ijms20081956. [PMID: 31013569 PMCID: PMC6514982 DOI: 10.3390/ijms20081956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
Congenital FXIII deficiency is a rare bleeding disorder in which mutations are detected in F13A1 and F13B genes that express the two subunits of coagulation FXIII, the catalytic FXIII-A, and protective FXIII-B. Mutations in FXIII-B subunit are considerably rarer compared to FXIII-A. Three mutations in the F13B gene have been reported on its structural disulfide bonds. In the present study, we investigate the structural and functional importance of all 20 structural disulfide bonds in FXIII-B subunit. All disulfide bonds were ablated by individually mutating one of its contributory cysteine’s, and these variants were transiently expressed in HEK293t cell lines. The expression products were studied for stability, secretion, the effect on oligomeric state, and on FXIII-A activation. The structural flexibility of these disulfide bonds was studied using classical MD simulation performed on a FXIII-B subunit monomer model. All 20 FXIII-B were found to be important for the secretion and stability of the protein since ablation of any of these led to a secretion deficit. However, the degree of effect that the disruption of disulfide bond had on the protein differed between individual disulfide bonds reflecting a functional hierarchy/diversity within these disulfide bonds.
Collapse
|
30
|
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
|
31
|
Wang H, Zhang L, Tao Y, Wang Z, Shen D, Dong H. Transmembrane Helices 2 and 3 Determine the Localization of Plasma Membrane Intrinsic Proteins in Eukaryotic Cells. FRONTIERS IN PLANT SCIENCE 2019; 10:1671. [PMID: 31998350 PMCID: PMC6966961 DOI: 10.3389/fpls.2019.01671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/27/2019] [Indexed: 05/12/2023]
Abstract
In plants, plasma membrane intrinsic protein (PIP) PIP1s and PIP2s mediate the transport of disparate substrates across plasma membranes (PMs), with a prerequisite that the proteins correctly localize to the PMs. While PIP2s can take correct localization by themselves in plant cells, PIP1s cannot unless aided by a specific PIP2. Here, we analyzed the localization of the Arabidopsis aquaporins, AtPIP1s, AtPIP2;4, and their mutants in yeast, Xenopus oocytes, and protoplasts of Arabidopsis. Most of AtPIP2;4 localized in the PM when expressed alone, whereas AtPIP1;1 failed to realize it in yeast and Xenopus oocytes. Switch of the transmembrane helix 2 (TM2) or TM3 from AtPIP1;1 to AtPIP2;4 disabled the latter's PM targeting activity. Surprisingly, a replacement of TM2 and TM3 of AtPIP1;1 with those of AtPIP2;4 created a PM-localized AtPIP1;1 mutant, 1;1Δ(TM2+TM3)/2;4(TM2+TM3), which could act as a water and hydrogen peroxide channel just like AtPIP2;4. A localization and function analysis on mutants of AtPIP1;2, AtPIP1;3, AtPIP1;4, and AtPIP1;5, with the same replaced TM2 and TM3 from AtPIP2;4, showed that these AtPIP1 variants could also localize in the PM spontaneously, thus playing an inherent role in transporting solutes. Sequential and structural analysis suggested that a hydrophilic residue and a defective LxxxA motif are modulators of PM localization of AtPIP1s. These results indicate that TM2 and TM3 are necessary and, more importantly, sufficient in AtPIP2 for its PM localization.
Collapse
Affiliation(s)
- Hao Wang
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, China
| | - Liyuan Zhang
- Department of Plant Pathology, Shandong Agricultural University, Taian, China
- State Key Laboratory of Crop Biology, Taian, China
| | - Yuan Tao
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, China
| | - Zuodong Wang
- Department of Plant Pathology, Shandong Agricultural University, Taian, China
- State Key Laboratory of Crop Biology, Taian, China
| | - Dan Shen
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Dan Shen, ; Hansong Dong,
| | - Hansong Dong
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, China
- Department of Plant Pathology, Shandong Agricultural University, Taian, China
- State Key Laboratory of Crop Biology, Taian, China
- *Correspondence: Dan Shen, ; Hansong Dong,
| |
Collapse
|
32
|
di Ronza A, Bajaj L, Sharma J, Sanagasetti D, Lotfi P, Adamski CJ, Collette J, Palmieri M, Amawi A, Popp L, Chang KT, Meschini MC, Leung HCE, Segatori L, Simonati A, Sifers RN, Santorelli FM, Sardiello M. CLN8 is an endoplasmic reticulum cargo receptor that regulates lysosome biogenesis. Nat Cell Biol 2018; 20:1370-1377. [PMID: 30397314 PMCID: PMC6277210 DOI: 10.1038/s41556-018-0228-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
Organelle biogenesis requires proper transport of proteins from their site of synthesis to their target subcellular compartment1-3. Lysosomal enzymes are synthesized in the endoplasmic reticulum (ER) and traffic through the Golgi complex before being transferred to the endolysosomal system4-6, but how they are transferred from the ER to the Golgi is unknown. Here, we show that ER-to-Golgi transfer of lysosomal enzymes requires CLN8, an ER-associated membrane protein whose loss of function leads to the lysosomal storage disorder, neuronal ceroid lipofuscinosis 8 (a type of Batten disease)7. ER-to-Golgi trafficking of CLN8 requires interaction with the COPII and COPI machineries via specific export and retrieval signals localized in the cytosolic carboxy terminus of CLN8. CLN8 deficiency leads to depletion of soluble enzymes in the lysosome, thus impairing lysosome biogenesis. Binding to lysosomal enzymes requires the second luminal loop of CLN8 and is abolished by some disease-causing mutations within this region. Our data establish an unanticipated example of an ER receptor serving the biogenesis of an organelle and indicate that impaired transport of lysosomal enzymes underlies Batten disease caused by mutations in CLN8.
Collapse
Affiliation(s)
- Alberto di Ronza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Lakshya Bajaj
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jaiprakash Sharma
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Deepthi Sanagasetti
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Carolyn Joy Adamski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - John Collette
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Michela Palmieri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Abdallah Amawi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Lauren Popp
- Departments of Bioengineering, Chemical and Biomolecular Engineering, and Biochemistry and Cell Biology, Rice University, Houston, TX, USA
| | - Kevin Tommy Chang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Biosciences, Rice University, Houston, TX, USA
| | - Maria Chiara Meschini
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Hon-Chiu Eastwood Leung
- Departments of Medicine, Pediatrics, and Molecular and Cellular Biology, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Laura Segatori
- Departments of Bioengineering, Chemical and Biomolecular Engineering, and Biochemistry and Cell Biology, Rice University, Houston, TX, USA
| | - Alessandro Simonati
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Richard Norman Sifers
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
33
|
Melero-Fernandez de Mera RM, Arasu UT, Kärnä R, Oikari S, Rilla K, Vigetti D, Passi A, Heldin P, Tammi MI, Deen AJ. Effects of mutations in the post-translational modification sites on the trafficking of hyaluronan synthase 2 (HAS2). Matrix Biol 2018; 80:85-103. [PMID: 30394292 DOI: 10.1016/j.matbio.2018.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 01/01/2023]
Abstract
Vesicular trafficking of hyaluronan synthases (HAS1-3) from endoplasmic reticulum (ER) through Golgi to plasma membrane (PM), and either back to endosomes and lysosomes, or out into extracellular vesicles, is important for their activities. We studied how post-translational modifications affect the trafficking of HAS2 by mutagenesis of the sites of ubiquitination (K190R), phosphorylation (T110A) and O-GlcNAcylation (S221A), using Dendra2- and EGFP-HAS2 transfected into COS1 cells. Confocal microscopy showed HAS2 wild type (wt) and its K190R and S221A mutants in ER, Golgi and extracellular vesicles, while the T110A mutant remained mostly in the ER. HA synthesis was reduced by S221A, while completely blocked by K190R and T110A. Cell-surface biotinylation indicated that T110A was absent from PM, while S221A was close to the level of wt, and K190R was increased in PM. TIRF microscopy analysis gave similar results. Rab10 silencing increased HA secretion by HAS2, likely by inhibiting endocytosis of the enzyme from PM, as reported before for HAS3. Green-to-red photo-conversion of Dendra2-HAS2 constructs suggested slower decay of K190R and S221A than HAS2 wt, while T110A was barely degraded at all. S221D and S221E, the phosphomimetic mutants of this site, decayed faster and blocked hyaluronan synthesis, suggesting alternative O-GlcNAc/-PO4 substitution to regulate the stability of the enzyme. Probing the role of dynamic O-GlcNAcylation at S221 by adding glucosamine increased the half-life of only HAS2 wt. The Dendra2·HAS2 disappearance from Golgi was slower for K190R. Of the two inactive constructs, K190R co-transfected with HAS2 wt suppressed, whereas T110A had no effect on HA synthesis. Interestingly, the HAS2-stimulated shedding of extracellular vesicles was dependent on HAS residence in PM but independent of HA synthesis. The results indicate that post-translational modifications control the trafficking of HAS2, and that trafficking is an integral part of the post-translational regulation of HAS2 activity.
Collapse
Affiliation(s)
| | - U T Arasu
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - R Kärnä
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - S Oikari
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - K Rilla
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - D Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - A Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - P Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - M I Tammi
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - A J Deen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
34
|
Aviram N, Schuldiner M. Targeting and translocation of proteins to the endoplasmic reticulum at a glance. J Cell Sci 2018; 130:4079-4085. [PMID: 29246967 DOI: 10.1242/jcs.204396] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The evolutionary emergence of organelles was a defining process in diversifying biochemical reactions within the cell and enabling multicellularity. However, compartmentalization also imposed a great challenge-the need to import proteins synthesized in the cytosol into their respective sites of function. For example, one-third of all genes encode for proteins that must be targeted and translocated into the endoplasmic reticulum (ER), which serves as the entry site to the majority of endomembrane compartments. Decades of research have set down the fundamental principles of how proteins get from the cytosol into the ER, and recent studies have brought forward new pathways and additional regulators enabling better definition of the rules governing substrate recognition. In this Cell Science at a Glance article and the accompanying poster, we give an overview of our current understanding of the multifaceted and regulated processes of protein targeting and translocation to the ER.
Collapse
Affiliation(s)
- Naama Aviram
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel 7610001
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel 7610001
| |
Collapse
|
35
|
Mackie TD, Kim BY, Subramanya AR, Bain DJ, O'Donnell AF, Welling PA, Brodsky JL. The endosomal trafficking factors CORVET and ESCRT suppress plasma membrane residence of the renal outer medullary potassium channel (ROMK). J Biol Chem 2018; 293:3201-3217. [PMID: 29311259 PMCID: PMC5836112 DOI: 10.1074/jbc.m117.819086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/02/2018] [Indexed: 11/06/2022] Open
Abstract
Protein trafficking can act as the primary regulatory mechanism for ion channels with high open probabilities, such as the renal outer medullary (ROMK) channel. ROMK, also known as Kir1.1 (KCNJ1), is the major route for potassium secretion into the pro-urine and plays an indispensable role in regulating serum potassium and urinary concentrations. However, the cellular machinery that regulates ROMK trafficking has not been fully defined. To identify regulators of the cell-surface population of ROMK, we expressed a pH-insensitive version of the channel in the budding yeast Saccharomyces cerevisiae We determined that ROMK primarily resides in the endoplasmic reticulum (ER), as it does in mammalian cells, and is subject to ER-associated degradation (ERAD). However, sufficient ROMK levels on the plasma membrane rescued growth on low-potassium medium of yeast cells lacking endogenous potassium channels. Next, we aimed to identify the biological pathways most important for ROMK regulation. Therefore, we used a synthetic genetic array to identify non-essential genes that reduce the plasma membrane pool of ROMK in potassium-sensitive yeast cells. Genes identified in this screen included several members of the endosomal complexes required for transport (ESCRT) and the class-C core vacuole/endosome tethering (CORVET) complexes. Mass spectroscopy analysis confirmed that yeast cells lacking an ESCRT component accumulate higher potassium concentrations. Moreover, silencing of ESCRT and CORVET components increased ROMK levels at the plasma membrane in HEK293 cells. Our results indicate that components of the post-endocytic pathway influence the cell-surface density of ROMK and establish that components in this pathway modulate channel activity.
Collapse
Affiliation(s)
| | - Bo-Young Kim
- the Department of Physiology, University of Maryland at Baltimore, Baltimore, Maryland 21201
| | - Arohan R Subramanya
- the Departments of Medicine and Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- the Medicine and Research Services, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania 15240, and
| | - Daniel J Bain
- Geology and Environmental Science, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Allyson F O'Donnell
- the Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282
| | - Paul A Welling
- the Department of Physiology, University of Maryland at Baltimore, Baltimore, Maryland 21201
| | | |
Collapse
|
36
|
Abstract
In eukaryotes, distinct transport vesicles functionally connect various intracellular compartments. These carriers mediate transport of membranes for the biogenesis and maintenance of organelles, secretion of cargo proteins and peptides, and uptake of cargo into the cell. Transport vesicles have distinct protein coats that assemble on a donor membrane where they can select cargo and curve the membrane to form a bud. A multitude of structural elements of coat proteins have been solved by X-ray crystallography. More recently, the architectures of the COPI and COPII coats were elucidated in context with their membrane by cryo-electron tomography. Here, we describe insights gained from the structures of these two coat lattices and discuss the resulting functional implications.
Collapse
Affiliation(s)
- Julien Béthune
- Heidelberg University Biochemistry Centre, 69120 Heidelberg, Germany; ,
| | - Felix T Wieland
- Heidelberg University Biochemistry Centre, 69120 Heidelberg, Germany; ,
| |
Collapse
|
37
|
Vanshylla K, Opazo F, Gronke K, Wienands J, Engels N. The extracellular membrane-proximal domain of membrane-bound IgE restricts B cell activation by limiting B cell antigen receptor surface expression. Eur J Immunol 2017; 48:441-453. [PMID: 29150831 DOI: 10.1002/eji.201747196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/03/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022]
Abstract
Immunoglobulin E (IgE) antibodies are key mediators of allergic reactions. Due to their potentially harmful anaphylactic properties, their production is tightly regulated. The membrane-bound isoform of IgE (mIgE), which is an integral component of the B cell antigen receptor, has been shown to be critical for the regulation of IgE responses in mice. In primate species including humans, mIgE can be expressed in two isoforms that are produced by alternative splicing of the primary ε Ig heavy chain transcript, and differ in the absence or presence of an extracellular membrane-proximal domain (EMPD) consisting of 52 amino acids. However, the function of the EMPD remains unclear. Here, we demonstrate that the EMPD restricts surface expression of mIgE-containing BCRs in human and murine B cells. The EMPD does not interfere with BCR assembly but acts as an autonomous endoplasmic reticulum retention domain. Limited surface expression of EMPD-containing mIgE-BCRs caused impaired activation of intracellular signaling cascades and hence represents a regulatory mechanism that may control the production of potentially anaphylactic IgE antibodies in primate species.
Collapse
Affiliation(s)
- Kanika Vanshylla
- Institute of Cellular & Molecular Immunology, Unversity Medical Center Göttingen, Göttingen, Germany
| | - Felipe Opazo
- Institute of Neuro- & Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Göttingen, Germany
| | - Konrad Gronke
- Institute of Cellular & Molecular Immunology, Unversity Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology, Unversity Medical Center Göttingen, Göttingen, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, Unversity Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
38
|
Geva Y, Crissman J, Arakel EC, Gómez-Navarro N, Chuartzman SG, Stahmer KR, Schwappach B, Miller EA, Schuldiner M. Two novel effectors of trafficking and maturation of the yeast plasma membrane H + -ATPase. Traffic 2017; 18:672-682. [PMID: 28727280 PMCID: PMC5607100 DOI: 10.1111/tra.12503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 11/28/2022]
Abstract
The endoplasmic reticulum (ER) is the entry site of proteins into the endomembrane system. Proteins exit the ER via coat protein II (COPII) vesicles in a selective manner, mediated either by direct interaction with the COPII coat or aided by cargo receptors. Despite the fundamental role of such receptors in protein sorting, only a few have been identified. To further define the machinery that packages secretory cargo and targets proteins from the ER to Golgi membranes, we used multiple systematic approaches, which revealed 2 uncharacterized proteins that mediate the trafficking and maturation of Pma1, the essential yeast plasma membrane proton ATPase. Ydl121c (Exp1) is an ER protein that binds Pma1, is packaged into COPII vesicles, and whose deletion causes ER retention of Pma1. Ykl077w (Psg1) physically interacts with Exp1 and can be found in the Golgi and coat protein I (COPI) vesicles but does not directly bind Pma1. Loss of Psg1 causes enhanced degradation of Pma1 in the vacuole. Our findings suggest that Exp1 is a Pma1 cargo receptor and that Psg1 aids Pma1 maturation in the Golgi or affects its retrieval. More generally our work shows the utility of high content screens in the identification of novel trafficking components.
Collapse
Affiliation(s)
- Yosef Geva
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jonathan Crissman
- Department of Biological Sciences, Columbia University, New York, NY
| | - Eric C Arakel
- Department of Molecular Biology, Universitätsmedizin Göttingen, Göttingen, Germany
| | | | - Silvia G Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Kyle R Stahmer
- Department of Biological Sciences, Columbia University, New York, NY
| | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Elizabeth A Miller
- Department of Biological Sciences, Columbia University, New York, NY.,MRC Laboratory of Molecular Biology, Cell Biology Division, Cambridge, UK
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
39
|
Sirkis DW, Aparicio RE, Schekman R. Neurodegeneration-associated mutant TREM2 proteins abortively cycle between the ER and ER-Golgi intermediate compartment. Mol Biol Cell 2017; 28:2723-2733. [PMID: 28768830 PMCID: PMC5620379 DOI: 10.1091/mbc.e17-06-0423] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 11/21/2022] Open
Abstract
Mutations in the microglial cell surface receptor TREM2 are associated with multiple forms of neurodegeneration. Several of these mutant forms of TREM2 were thought to be retained in the endoplasmic reticulum (ER), but careful analysis reveals that they engage in an abortive cycling pathway between the ER and ER–Golgi intermediate compartment. Triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane protein expressed on microglia within the brain. Several rare mutations in TREM2 cause an early-onset form of neurodegeneration when inherited homozygously. Here we investigate how these mutations affect the intracellular transport of TREM2. We find that most pathogenic TREM2 mutant proteins fail to undergo normal maturation in the Golgi complex and show markedly reduced cell-surface expression. Prior research has suggested that two such mutants are retained in the endoplasmic reticulum (ER), but we find, using a cell-free coat protein complex II (COPII) vesicle budding reaction, that mutant TREM2 is exported efficiently from the ER. In addition, mutant TREM2 becomes sensitive to cleavage by endoglycosidase D under conditions that inhibit recycling to the ER, indicating that it normally reaches a post-ER compartment. Maturation-defective TREM2 mutants are also efficiently bound by a lectin that recognizes O-glycans added in the ER–Golgi intermediate compartment (ERGIC) and cis-Golgi cisterna. Finally, mutant TREM2 accumulates in the ERGIC in cells depleted of COPI. These results indicate that efficient ER export is not sufficient to enable normal cell-surface expression of TREM2. Moreover, our findings suggest that the ERGIC may play an underappreciated role as a quality-control center for mutant and/or malformed membrane proteins.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Renan E Aparicio
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
40
|
Protein quality control at the endoplasmic reticulum. Essays Biochem 2017; 60:227-235. [PMID: 27744338 DOI: 10.1042/ebc20160003] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/31/2016] [Indexed: 12/22/2022]
Abstract
The ER (endoplasmic reticulum) is the protein folding 'factory' of the secretory pathway. Virtually all proteins destined for the plasma membrane, the extracellular space or other secretory compartments undergo folding and maturation within the ER. The ER hosts a unique PQC (protein quality control) system that allows specialized modifications such as glycosylation and disulfide bond formation essential for the correct folding and function of many secretory proteins. It is also the major checkpoint for misfolded or aggregation-prone proteins that may be toxic to the cell or extracellular environment. A failure of this system, due to aging or other factors, has therefore been implicated in a number of serious human diseases. In this article, we discuss several key features of ER PQC that maintain the health of the cellular secretome.
Collapse
|
41
|
Ferguson S, Steyer AM, Mayhew TM, Schwab Y, Lucocq JM. Quantifying Golgi structure using EM: combining volume-SEM and stereology for higher throughput. Histochem Cell Biol 2017; 147:653-669. [PMID: 28429122 PMCID: PMC5429891 DOI: 10.1007/s00418-017-1564-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2017] [Indexed: 12/28/2022]
Abstract
Investigating organelles such as the Golgi complex depends increasingly on high-throughput quantitative morphological analyses from multiple experimental or genetic conditions. Light microscopy (LM) has been an effective tool for screening but fails to reveal fine details of Golgi structures such as vesicles, tubules and cisternae. Electron microscopy (EM) has sufficient resolution but traditional transmission EM (TEM) methods are slow and inefficient. Newer volume scanning EM (volume-SEM) methods now have the potential to speed up 3D analysis by automated sectioning and imaging. However, they produce large arrays of sections and/or images, which require labour-intensive 3D reconstruction for quantitation on limited cell numbers. Here, we show that the information storage, digital waste and workload involved in using volume-SEM can be reduced substantially using sampling-based stereology. Using the Golgi as an example, we describe how Golgi populations can be sensed quantitatively using single random slices and how accurate quantitative structural data on Golgi organelles of individual cells can be obtained using only 5–10 sections/images taken from a volume-SEM series (thereby sensing population parameters and cell–cell variability). The approach will be useful in techniques such as correlative LM and EM (CLEM) where small samples of cells are treated and where there may be variable responses. For Golgi study, we outline a series of stereological estimators that are suited to these analyses and suggest workflows, which have the potential to enhance the speed and relevance of data acquisition in volume-SEM.
Collapse
Affiliation(s)
- Sophie Ferguson
- Structural Cell Biology Group, School of Medicine, University of St Andrews, North Haugh, Fife, KY16 9TF, Scotland, UK
| | - Anna M Steyer
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Terry M Mayhew
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - John Milton Lucocq
- Structural Cell Biology Group, School of Medicine, University of St Andrews, North Haugh, Fife, KY16 9TF, Scotland, UK.
| |
Collapse
|
42
|
Liu L, Tian J, Lu C, Chen X, Fu Y, Xu B, Zhu C, Sun Y, Zhang Y, Zhao Y, Li Y. Electrophysiological Characteristics of the LQT2 Syndrome Mutation KCNH2-G572S and Regulation by Accessory Protein KCNE2. Front Physiol 2016; 7:650. [PMID: 28082916 PMCID: PMC5187237 DOI: 10.3389/fphys.2016.00650] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/12/2016] [Indexed: 01/18/2023] Open
Abstract
Mutations in hERG cause long QT syndrome type 2 which is characterized by a prolonged QT interval on electrocardiogram and predisposition to life-threatening ventricular tachyarrhythmia, syncope, and sudden death. hERG-G572S induces trafficking defects of hERG channel protein from Golgi to the plasma membrane and results in a dominant negative suppression of hERG current density. As an accessory β subunit, KCNE2 promotes hERG migration from Golgi to cellular membrane. In this study, we investigated the rescue effect of KCNE2 in a G572S mutation of hERG. Transfection was performed into HEK293 cells. Patch clamp technique, western blotting analyses and confocal microscopic examination were used. Results showed that KCNE2 had a significantly enhanced effect on G572S mutation current. The increase of current was largest at KCNH2:KCNE2 of 1:3. Confocal images showed co-expressing G572S and KCNE2 could cause a substantial up-regulated membrane protein (155 kDa) expression. Expression of membrane protein accumulated markedly with increasing ratio of KCNH2:KCNE2. G572S defective mutant could be restored by both KCNE2 and lower temperature (27°C), which suggested that the lower temperature could be the favorable circumstances for the rescue function of KCNE2. In this study, we successfully set up “the action potential” on the HEK 293 cells by genetically engineered to express Kir2.1, Nav1.5, and Kv11.1, wherein on reaching over an excitation threshold by current injection. The results suggested that KCNE2 could shorten action potential duration which was prolonged by G572S. These findings described electrophysiological characteristics of the LQT2 syndrome mutation KCNH2-G572S and regulation by accessory protein KCNE2, and provided a clue about LQT2 and relative rescue mechanism.
Collapse
Affiliation(s)
- Li Liu
- Department of Cardiology, General Hospital of People's Liberation ArmyBeijing, China; The Third Department of Internal Medicine, Beijing Municipal Corps Hospital of Chinese People's Armed Police ForceBeijing, China
| | - Jinwen Tian
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Caiyi Lu
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Xi Chen
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Yicheng Fu
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Bin Xu
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Chao Zhu
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Yanmei Sun
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Yu Zhang
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Ying Zhao
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| | - Yang Li
- Department of Cardiology, General Hospital of People's Liberation Army Beijing, China
| |
Collapse
|
43
|
Gomez-Navarro N, Miller E. Protein sorting at the ER-Golgi interface. J Cell Biol 2016; 215:769-778. [PMID: 27903609 PMCID: PMC5166505 DOI: 10.1083/jcb.201610031] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/02/2016] [Accepted: 11/17/2016] [Indexed: 01/01/2023] Open
Abstract
In this review, Gomez-Navarro and Miller summarize the principles of cargo sorting by the vesicle traffic machinery and consider the diverse mechanisms by which cargo proteins are selected and captured into different transport vesicles. Protein traffic is of critical importance for normal cellular physiology. In eukaryotes, spherical transport vesicles move proteins and lipids from one internal membrane-bound compartment to another within the secretory pathway. The process of directing each individual protein to a specific destination (known as protein sorting) is a crucial event that is intrinsically linked to vesicle biogenesis. In this review, we summarize the principles of cargo sorting by the vesicle traffic machinery and consider the diverse mechanisms by which cargo proteins are selected and captured into different transport vesicles. We focus on the first two compartments of the secretory pathway: the endoplasmic reticulum and Golgi. We provide an overview of the complexity and diversity of cargo adaptor function and regulation, focusing on recent mechanistic discoveries that have revealed insight into protein sorting in cells.
Collapse
Affiliation(s)
- Natalia Gomez-Navarro
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Elizabeth Miller
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| |
Collapse
|
44
|
Lee H, Noh H, Mun J, Gu C, Sever S, Park S. Anks1a regulates COPII-mediated anterograde transport of receptor tyrosine kinases critical for tumorigenesis. Nat Commun 2016; 7:12799. [PMID: 27619642 PMCID: PMC5027278 DOI: 10.1038/ncomms12799] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023] Open
Abstract
ErbB2 signalling, which is amplified by EphA2 binding, is an important therapeutic target for breast cancer. Despite the importance of the EphA2/ErbB2 complex in promoting breast tumorigenesis, the mechanism by which these receptor tyrosine kinases (RTKs) are exported from the endoplasmic reticulum (ER) remains poorly understood. Here we report that the PTB adaptor Anks1a is specifically localized to the ER on its own serine phosphorylation. Once there, Anks1a acts as an important regulator of COPII-mediated EphA2 ER export. The Anks1a ankyrin repeat domain binds EphA2 and causes it to accumulate at sites of ER exit. Simultaneously, the Anks1a PTB domain binds Sec23. This induces internalization of EphA2 via COPII vesicles, while Anks1a remains behind on the ER membrane. EphA2 also binds ErbB2 in the ER and seems to load ErbB2 into growing COPII carriers. Together, our study reveals a novel mechanism that regulates the loading of RTKs into COPII vesicles. EphA2/ErbB2 complex is important in promoting breast cancer but the mechanism by which these receptor tyrosine kinases are exported from the endoplasmic reticulum is unknown. Here the authors show that Anks1a acts as a cargo adaptor in sorting EphA2 into COPII vesicles, thus modulating the surface level of EphA2.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Biological Science, Sookmyung Women's University, Chungpa-ro 47gil 100, Yongsan-gu, Seoul 140-742, Korea
| | - Hyuna Noh
- Department of Biological Science, Sookmyung Women's University, Chungpa-ro 47gil 100, Yongsan-gu, Seoul 140-742, Korea
| | - Jiyoung Mun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam-Si, Gyeonggi-Do 13135, Korea
| | - Changkyu Gu
- Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Sanja Sever
- Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Soochul Park
- Department of Biological Science, Sookmyung Women's University, Chungpa-ro 47gil 100, Yongsan-gu, Seoul 140-742, Korea
| |
Collapse
|
45
|
Staudt C, Gilis F, Boonen M, Jadot M. Molecular determinants that mediate the sorting of human ATG9A from the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2299-310. [PMID: 27316455 DOI: 10.1016/j.bbamcr.2016.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/07/2016] [Accepted: 06/14/2016] [Indexed: 11/15/2022]
Abstract
ATG9A is a multispanning membrane protein required for autophagosome formation. Under basal conditions, neosynthesized ATG9A proteins travel to the Golgi apparatus and cycle between the trans-Golgi network and endosomes. In the present work, we searched for molecular determinants involved in the subcellular trafficking of human ATG9A in HeLa cells using sequential deletions and point mutations. Deletion of amino acids L(340) to L(354) resulted in the retention of ATG9A in the endoplasmic reticulum. In addition, we found that substitution of the L(711)YM(713) sequence (located in the C-terminal region of ATG9A) by alanine residues severely impaired its transport through the Golgi apparatus. This defect could be corrected by oligomerization of the mutant protein with co-transfected wild-type ATG9A, suggesting that ATG9A oligomerization may help its sorting through biosynthetic compartments. Lastly, the study of the consequences of the LYM/AAA mutation on the intracellular trafficking of ATG9A highlighted that some newly synthesized ATG9A can bypass the Golgi apparatus to reach the plasma membrane. Taken together, these findings provide new insights into the intracellular pathways followed by ATG9A to reach different subcellular compartments, and into the intramolecular determinants that drive the sorting of this protein.
Collapse
Affiliation(s)
- Catherine Staudt
- URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, Belgium
| | - Florentine Gilis
- URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, Belgium
| | - Marielle Boonen
- URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, Belgium
| | - Michel Jadot
- URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, Belgium.
| |
Collapse
|
46
|
Thinon E, Morales-Sanfrutos J, Mann DJ, Tate EW. N-Myristoyltransferase Inhibition Induces ER-Stress, Cell Cycle Arrest, and Apoptosis in Cancer Cells. ACS Chem Biol 2016; 11:2165-76. [PMID: 27267252 PMCID: PMC5077176 DOI: 10.1021/acschembio.6b00371] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022]
Abstract
N-Myristoyltransferase (NMT) covalently attaches a C14 fatty acid to the N-terminal glycine of proteins and has been proposed as a therapeutic target in cancer. We have recently shown that selective NMT inhibition leads to dose-responsive loss of N-myristoylation on more than 100 protein targets in cells, and cytotoxicity in cancer cells. N-myristoylation lies upstream of multiple pro-proliferative and oncogenic pathways, but to date the complex substrate specificity of NMT has limited determination of which diseases are most likely to respond to a selective NMT inhibitor. We describe here the phenotype of NMT inhibition in HeLa cells and show that cells die through apoptosis following or concurrent with accumulation in the G1 phase. We used quantitative proteomics to map protein expression changes for more than 2700 proteins in response to treatment with an NMT inhibitor in HeLa cells and observed down-regulation of proteins involved in cell cycle regulation and up-regulation of proteins involved in the endoplasmic reticulum stress and unfolded protein response, with similar results in breast (MCF-7, MDA-MB-231) and colon (HCT116) cancer cell lines. This study describes the cellular response to NMT inhibition at the proteome level and provides a starting point for selective targeting of specific diseases with NMT inhibitors, potentially in combination with other targeted agents.
Collapse
Affiliation(s)
- Emmanuelle Thinon
- Department
of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
- Department
of Life Sciences, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
| | - Julia Morales-Sanfrutos
- Department
of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
| | - David J. Mann
- Department
of Life Sciences, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
- Institute
of Chemical Biology, Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
| | - Edward W. Tate
- Department
of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
- Institute
of Chemical Biology, Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, United Kingdom
| |
Collapse
|
47
|
Stijnen P, Ramos-Molina B, O'Rahilly S, Creemers JWM. PCSK1 Mutations and Human Endocrinopathies: From Obesity to Gastrointestinal Disorders. Endocr Rev 2016; 37:347-71. [PMID: 27187081 DOI: 10.1210/er.2015-1117] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prohormone convertase 1/3, encoded by the PCSK1 gene, is a serine endoprotease that is involved in the processing of a variety of proneuropeptides and prohormones. Humans who are homozygous or compound heterozygous for loss-of-function mutations in PCSK1 exhibit a variable and pleiotropic syndrome consisting of some or all of the following: obesity, malabsorptive diarrhea, hypogonadotropic hypogonadism, altered thyroid and adrenal function, and impaired regulation of plasma glucose levels in association with elevated circulating proinsulin-to-insulin ratio. Recently, more common variants in the PCSK1 gene have been found to be associated with alterations in body mass index, increased circulating proinsulin levels, and defects in glucose homeostasis. This review provides an overview of the endocrinopathies and other disorders observed in prohormone convertase 1/3-deficient patients, discusses the possible biochemical basis for these manifestations of the disease, and proposes a model whereby certain missense mutations in PCSK1 may result in proteins with a dominant negative action.
Collapse
Affiliation(s)
- Pieter Stijnen
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Bruno Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephen O'Rahilly
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - John W M Creemers
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
48
|
Abstract
Transport of newly synthesized proteins from the endoplasmic reticulum (ER) to the Golgi complex is highly selective. As a general rule, such transport is limited to soluble and membrane-associated secretory proteins that have reached properly folded and assembled conformations. To secure the efficiency, fidelity, and control of this crucial transport step, cells use a combination of mechanisms. The mechanisms are based on selective retention of proteins in the ER to prevent uptake into transport vesicles, on selective capture of proteins in COPII carrier vesicles, on inclusion of proteins in these vesicles by default as part of fluid and membrane bulk flow, and on selective retrieval of proteins from post-ER compartments by retrograde vesicle transport.
Collapse
Affiliation(s)
- Charles Barlowe
- Biochemistry Department, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755;
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich CH-8093, Switzerland
| |
Collapse
|
49
|
Borgese N. Getting membrane proteins on and off the shuttle bus between the endoplasmic reticulum and the Golgi complex. J Cell Sci 2016; 129:1537-45. [PMID: 27029344 DOI: 10.1242/jcs.183335] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Secretory proteins exit the endoplasmic reticulum (ER) in coat protein complex II (COPII)-coated vesicles and then progress through the Golgi complex before delivery to their final destination. Soluble cargo can be recruited to ER exit sites by signal-mediated processes (cargo capture) or by bulk flow. For membrane proteins, a third mechanism, based on the interaction of their transmembrane domain (TMD) with lipid microdomains, must also be considered. In this Commentary, I review evidence in favor of the idea that partitioning of TMDs into bilayer domains that are endowed with distinct physico-chemical properties plays a pivotal role in the transport of membrane proteins within the early secretory pathway. The combination of such self-organizational phenomena with canonical intermolecular interactions is most likely to control the release of membrane proteins from the ER into the secretory pathway.
Collapse
Affiliation(s)
- Nica Borgese
- CNR Institute of Neuroscience, Milan 20129, Italy
| |
Collapse
|
50
|
Stijnen P, Brouwers B, Dirkx E, Ramos-Molina B, Van Lommel L, Schuit F, Thorrez L, Declercq J, Creemers JWM. Endoplasmic reticulum-associated degradation of the mouse PC1/3-N222D hypomorph and human PCSK1 mutations contributes to obesity. Int J Obes (Lond) 2016; 40:973-81. [PMID: 26786350 DOI: 10.1038/ijo.2016.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 12/03/2015] [Accepted: 12/22/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND The proprotein convertase 1/3 (PC1/3), encoded by proprotein convertase subtilisin/kexin type 1 (PCSK1), cleaves and hence activates several orexigenic and anorexigenic proproteins. Congenital inactivation of PCSK1 leads to obesity in human but not in mice. However, a mouse model harboring the hypomorphic mutation N222D is obese. It is not clear why the mouse models differ in phenotype. METHODS Gene expression analysis was performed with pancreatic islets from Pcsk1(N222D/N222D) mice. Subsequently, biosynthesis, maturation, degradation and activity were studied in islets, pituitary, hypothalamus and cell lines. Coimmunoprecipitation of PC1/3-N222D and human PC1/3 variants associated with obesity with the endoplasmic reticulum (ER) chaperone BiP was studied in cell lines. RESULTS Gene expression analysis of islets of Pcsk1(N222D/N222D) mice showed enrichment of gene sets related to the proteasome and the unfolded protein response. Steady-state levels of PC1/3-N222D and in particular the carboxy-terminally processed form were strongly reduced in islets, pituitary and hypothalamus. However, impairment of substrate cleavage was tissue dependent. Proinsulin processing was drastically reduced, while processing of proopiomelanocortin (POMC) to adrenocorticotropic hormone (ACTH) in pituitary was only mildly impaired. Growth hormone expression and IGF-1 levels were normal, indicating near-normal processing of hypothalamic proGHRH. PC1/3-N222D binds to BiP and is rapidly degraded by the proteasome. Analysis of human PC1/3 obesity-associated mutations showed increased binding to BiP and prolonged intracellular retention for all investigated mutations, in particular for PC1/3-T175M, PC1/3-G226R and PC1/3-G593R. CONCLUSIONS This study demonstrates that the hypomorphic mutation in Pcsk1(N222D) mice has an effect on catalytic activity in pancreatic islets, pituitary and hypothalamus. Reduced substrate processing activity in Pcsk1(N222D/N222D) mice is due to enhanced degradation in addition to reduced catalytic activity of the mutant. PC1/3-N222D binds to BiP, suggesting impaired folding and reduced stability. Enhanced BiP binding is also observed in several human obesity-associated PC1/3 variants, suggesting a common mechanism.
Collapse
Affiliation(s)
- P Stijnen
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - B Brouwers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - E Dirkx
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - B Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - L Van Lommel
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - F Schuit
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - L Thorrez
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - J Declercq
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - J W M Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|