1
|
Sharma R, Dey Das K, Srinivasula SM. EGF-mediated Golgi dynamics and cell migration require CARP2. Cell Rep 2024; 43:114896. [PMID: 39441718 DOI: 10.1016/j.celrep.2024.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/21/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
In mammalian cells, the Golgi exists in ribbon architecture-individual stacks laterally linked to each other by tubular structures. Golgi architecture changes dynamically to cater to cellular needs. Loss of architecture is linked with pathological conditions like cancer and neurodegeneration. Not much is known about the regulators of Golgi dynamics. Here, we demonstrate that CARP2 (caspase-8- and caspase-10-associated RING-containing protein 2), an endosomal ubiquitin ligase and a known regulator of cell migration, modulates Golgi dynamics. Epidermal growth factor (EGF) treatment modestly increases CARP2 protein and disperses Golgi. An exogenous supply of CARP2 also leads to Golgi dispersal. Conversely, Golgi remains intact in CARP2 knockout (KO) cells upon EGF treatment. CARP2 variants defective in either endosomal association or ligase activity are unable to affect Golgi dispersal. Importantly, CARP2 targets Golgin45 for ubiquitination and degradation in EGF-stimulated cells. Collectively, our findings unravel the existence of crosstalk between endosomal ubiquitin signaling and Golgi dynamics.
Collapse
Affiliation(s)
- Rahul Sharma
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Srinivasa M Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India.
| |
Collapse
|
2
|
Hashimoto Y, Kawade H, Bao W, Morii S, Nakano M, Nagae M, Murakami R, Tokoro Y, Nakashima M, Cai Z, Isaji T, Gu J, Nakajima K, Kizuka Y. The K346T mutant of GnT-III bearing weak in vitro and potent intracellular activity. Biochim Biophys Acta Gen Subj 2024; 1868:130663. [PMID: 38936637 DOI: 10.1016/j.bbagen.2024.130663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND N-Acetylglucosaminyltransferase-III (GnT-III, also designated MGAT3) catalyzes the formation of a specific N-glycan branch, bisecting GlcNAc, in the Golgi apparatus. Bisecting GlcNAc is a key residue that suppresses N-glycan maturation and is associated with the pathogenesis of cancer and Alzheimer's disease. However, it remains unclear how GnT-III recognizes its substrates and how GnT-III activity is regulated in cells. METHODS Using AlphaFold2 and structural comparisons, we predicted the key amino acid residues in GnT-III that interact with substrates in the catalytic pocket. We also performed in vitro activity assay, lectin blotting analysis and N-glycomic analysis using point mutants to assess their activity. RESULTS Our data suggested that E320 of human GnT-III is the catalytic center. More interestingly, we found a unique mutant, K346T, that exhibited lower in vitro activity and higher intracellular activity than wild-type GnT-III. The enzyme assays using various substrates showed that the substrate specificity of K346T was unchanged, whereas cycloheximide chase experiments revealed that the K346T mutant has a slightly shorter half-life, suggesting that the mutant is unstable possibly due to a partial misfolding. Furthermore, TurboID-based proximity labeling showed that the localization of the K346T mutant is shifted slightly to the cis side of the Golgi, probably allowing for prior action to competing galactosyltransferases. CONCLUSIONS The slight difference in K346T localization may be responsible for the higher biosynthetic activity despite the reduced activity. GENERAL SIGNIFICANCE Our findings underscore the importance of fine intra-Golgi localization and reaction orders of glycosyltransferases for the biosynthesis of complex glycan structures in cells.
Collapse
Affiliation(s)
- Yuta Hashimoto
- Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan
| | - Haruka Kawade
- Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan
| | - WanXue Bao
- Glyco-Biochemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Sayaka Morii
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima 739-8530, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima 739-8530, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita 565-0871, Japan
| | - Reiko Murakami
- Glycoanalytical Chemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Yuko Tokoro
- Glyco-Biochemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Misaki Nakashima
- Glyco-Biochemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Zixuan Cai
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Kazuki Nakajima
- Glycoanalytical Chemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Yasuhiko Kizuka
- Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan; Glyco-Biochemistry Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan.
| |
Collapse
|
3
|
Mahanty S, Bergam P, Belapurkar V, Eluvathingal L, Gupta N, Goud B, Nair D, Raposo G, Setty SRG. Biogenesis of specialized lysosomes in differentiated keratinocytes relies on close apposition with the Golgi apparatus. Cell Death Dis 2024; 15:496. [PMID: 38992005 PMCID: PMC11239851 DOI: 10.1038/s41419-024-06710-w] [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: 01/02/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 07/13/2024]
Abstract
Intracellular organelles support cellular physiology in diverse conditions. In the skin, epidermal keratinocytes undergo differentiation with gradual changes in cellular physiology, accompanying remodeling of lysosomes and the Golgi apparatus. However, it was not known whether changes in Golgi and lysosome morphology and their redistribution were linked. Here, we show that disassembled Golgi is distributed in close physical apposition to lysosomes in differentiated keratinocytes. This atypical localization requires the Golgi tethering protein GRASP65, which is associated with both the Golgi and lysosome membranes. Depletion of GRASP65 results in the loss of Golgi-lysosome apposition and the malformation of lysosomes, defined by their aberrant morphology, size, and function. Surprisingly, a trans-Golgi enzyme and secretory Golgi cargoes are extensively localized to the lysosome lumen and secreted to the cell surface, contributing to total protein secretion of differentiated keratinocytes but not in proliferative precursors, indicating that lysosomes acquire specialization during differentiation. We further demonstrate that the secretory function of the Golgi apparatus is critical to maintain keratinocyte lysosomes. Our study uncovers a novel form of Golgi-lysosome cross-talk and its role in maintaining specialized secretory lysosomes in differentiated keratinocytes.
Collapse
Affiliation(s)
- Sarmistha Mahanty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India.
| | - Ptissam Bergam
- Institut Curie, PSL Research University, CNRS, UMR 144, Structure and Membrane Compartments, F-75005, Paris, France
| | - Vivek Belapurkar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | | | - Nikita Gupta
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d'Ulm, F-75005, Paris, France
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR 144, Structure and Membrane Compartments, F-75005, Paris, France
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
4
|
Zhang JZ, Ong SE, Baker D, Maly DJ. Single-cell signaling analysis reveals that Major Vault Protein facilitates RasG12C inhibitor resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560617. [PMID: 37873412 PMCID: PMC10592919 DOI: 10.1101/2023.10.02.560617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Recently developed covalent inhibitors for RasG12C provide the first pharmacological tools to target mutant Ras-driven cancers. However, the rapid development of resistance to current clinical Ras G12C inhibitors is common. Presumably, a subpopulation of RasG12C-expressing cells adapt their signaling to evade these inhibitors and the mechanisms for this phenomenon are unclear due to the lack of tools that can measure signaling with single-cell resolution. Here, we utilized recently developed Ras sensors to profile the environment of active Ras and to measure the activity of endogenous Ras in order to pair structure (Ras signalosome) to function (Ras activity), respectively, at a single-cell level. With this approach, we identified a subpopulation of KRasG12C cells treated with RasG12C-GDP inhibitors underwent oncogenic signaling and metabolic changes driven by WT Ras at the golgi and mutant Ras at the mitochondria, respectively. Our Ras sensors identified Major Vault Protein (MVP) as a mediator of Ras activation at both compartments by scaffolding Ras signaling pathway components and metabolite channels. We found that recently developed RasG12C-GTP inhibitors also led to MVP-mediated WT Ras signaling at the golgi, demonstrating that this a general mechanism RasG12C inhibitor resistance. Overall, single-cell analysis of structure-function relationships enabled the discovery of a RasG12C inhibitor-resistant subpopulation driven by MVP, providing insight into the complex and heterogenous rewiring occurring during drug resistance in cancer.
Collapse
Affiliation(s)
- Jason Z. Zhang
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, United States
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
- Institute for Protein Design, University of Washington, Seattle, Washington 98195, United States
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, United States
| | - Dustin J. Maly
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
5
|
Sugiura K, Kawai Y, Yamamoto A, Yoshioka H, Kiyohara Y, Iida A, Ozawa Y, Nishikawa M, Miura N, Hanamatsu H, Furukawa JI, Shinohara Y. Exposure to brefeldin A induces unusual expression of hybrid- and complex-type free N-glycans in HepG2 cells. Biochim Biophys Acta Gen Subj 2023; 1867:130331. [PMID: 36804277 DOI: 10.1016/j.bbagen.2023.130331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023]
Abstract
This study determined the effect of brefeldin A (BFA) on the free N-glycomic profile of HepG2 cells to better understand the effect of blocking intracellular vesicle formation and transport of proteins from the endoplasmic reticulum to the Golgi apparatus. A series of exoglycosidase- and endoglycosidase-assisted analyses clarified the complex nature of altered glycomic profiles. A key feature of BFA-mediated alterations in Gn2-type glycans was the expression of unusual hybrid-, monoantennary- and complex-type free N-glycans (FNGs). BFA-mediated alterations in Gn1-type glycans were characterized by the expression of unusual hybrid- and monoantennary-FNGs, without significant expression of complex-type FNGs. A time course analysis revealed that sialylated hybrid- and complex-type Gn2-type FNGs were generated later than asialo-Gn2-type FNGs, and the expression profiles of Gn2-type FNGs and N-glycans were found to be similar, suggesting that the metabolic flux of FNGs is the same as that of protein-bound N-glycans. Subcellular glycomic analysis revealed that almost all FNGs were detected in the cytoplasmic extracts. Our data suggest that hybrid-, monoantennary- and complex-type Gn2-type FNGs were cleaved from glycoproteins in the cytosol by cytosolic PNGase, and subsequently digested by cytosolic endo-β-N-acetylglucosaminidase (ENGase) to generate Gn1-type FNGs. The substrate specificity of ENGase explains the limited expression of complex Gn1 type FNGs.
Collapse
Affiliation(s)
- Kanako Sugiura
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Yuho Kawai
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Arisa Yamamoto
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Hiroki Yoshioka
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
| | - Yuika Kiyohara
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Ayaka Iida
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Yurika Ozawa
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Mai Nishikawa
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Nobuaki Miura
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Hisatoshi Hanamatsu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita21, Nishi11, Kita-ku, Sapporo 001-0021, Japan
| | - Jun-Ichi Furukawa
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita21, Nishi11, Kita-ku, Sapporo 001-0021, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya 464-8601, Japan
| | - Yasuro Shinohara
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan; Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, Nagoya 463-8521, Japan.
| |
Collapse
|
6
|
Kotidis P, Donini R, Arnsdorf J, Hansen AH, Voldborg BGR, Chiang AWT, Haslam SM, Betenbaugh M, Jimenez Del Val I, Lewis NE, Krambeck F, Kontoravdi C. CHOGlycoNET: Comprehensive glycosylation reaction network for CHO cells. Metab Eng 2023; 76:87-96. [PMID: 36610518 PMCID: PMC11132536 DOI: 10.1016/j.ymben.2022.12.009] [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: 03/07/2022] [Revised: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023]
Abstract
Chinese hamster ovary (CHO) cells are extensively used for the production of glycoprotein therapeutics proteins, for which N-linked glycans are a critical quality attribute due to their influence on activity and immunogenicity. Manipulation of protein glycosylation is commonly achieved through cell or process engineering, which are often guided by mathematical models. However, each study considers a unique glycosylation reaction network that is tailored around the cell line and product at hand. Herein, we use 200 glycan datasets for both recombinantly produced and native proteins from different CHO cell lines to reconstruct a comprehensive reaction network, CHOGlycoNET, based on the individual minimal reaction networks describing each dataset. CHOGlycoNET is used to investigate the distribution of mannosidase and glycosyltransferase enzymes in the Golgi apparatus and identify key network reactions using machine learning and dimensionality reduction techniques. CHOGlycoNET can be used for accelerating glycomodel development and predicting the effect of glycoengineering strategies. Finally, CHOGlycoNET is wrapped in a SBML file to be used as a standalone model or in combination with CHO cell genome scale models.
Collapse
Affiliation(s)
- Pavlos Kotidis
- Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK.
| | - Roberto Donini
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Johnny Arnsdorf
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anders Holmgaard Hansen
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Bjørn Gunnar Rude Voldborg
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, CA, 92093, USA
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Michael Betenbaugh
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, CA, 92093, USA; Department of Bioengineering, University of California, San Diego, CA, 92093, USA
| | | | - Cleo Kontoravdi
- Sargent Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
7
|
Regulation of protein secretion through chemical regulation of endoplasmic reticulum retention signal cleavage. Nat Commun 2022; 13:1323. [PMID: 35260576 PMCID: PMC8904541 DOI: 10.1038/s41467-022-28971-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Secreted proteins, such as hormones or cytokines, are key mediators in multicellular organisms. Response of protein secretion based on transcriptional control is rather slow, as it requires transcription, translation and transport from the endoplasmic reticulum (ER) to the plasma membrane via the conventional protein secretion (CPS) pathway. An alternative regulation to provide faster response would be valuable. Here we present two genetically encoded orthogonal regulatory secretion systems, which rely on the retention of pre-synthesized proteins on the ER membrane (membER, released by a cytosolic protease) or inside the ER lumen (lumER, released by an ER-luminal protease), respectively, and their release by the chemical signal-regulated proteolytic removal of an ER-retention signal, without triggering ER stress due to protein aggregates. Design of orthogonal chemically-regulated split proteases enables the combination of signals into logic functions. Its application was demonstrated on a chemically regulated therapeutic protein secretion and regulated membrane translocation of a chimeric antigen receptor (CAR) targeting cancer antigen. Regulation of the ER escape represents a platform for the design of fast-responsive and tightly-controlled modular and scalable protein secretion system for mammalian cells. Secreted proteins, such as hormones or cytokines, are key mediators in multicellular organisms. Here the authors present two genetically encoded orthogonal regulatory secretion systems that enables inducible protein release and construction of logic gates.
Collapse
|
8
|
Yang H, Zhang F, Long H, Lin Y, Liao J, Xia H, Huang K. IFT20 Mediates the Transport of Cell Migration Regulators From the Trans-Golgi Network to the Plasma Membrane in Breast Cancer Cells. Front Cell Dev Biol 2021; 9:632198. [PMID: 33748116 PMCID: PMC7968458 DOI: 10.3389/fcell.2021.632198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/05/2021] [Indexed: 11/24/2022] Open
Abstract
IFT20 is a subunit of the intraflagellar transport (IFT) system essential for the formation and function of cilia. Besides predominant research in the cilia field, some IFT subunits perform extraciliary roles in non-ciliated cancer cells. However, the specific roles of IFT subunits in tumorigenesis remain unknown. Here, we found that knockout of IFT20 in mouse breast cancer cells lacking primary cilia promoted epithelial mesenchymal transitions (EMTs), active lamellipodia formation, and cell migration. IFT20 localized at the trans-Golgi and trans-Golgi network (TGN), and displayed vesicular co-distributions with Rab8a, the marker of TGN-to-plasma membrane vesicular trafficking. Proximity-dependent biotin identification (BioID) and colocalization analyzes showed that Numb and Ctnnal1, whose depletion promoted cell migration, co-localized with IFT20 at the trans-Golgi/TGN or intracellular transport vesicles. Furthermore, Strep-Tactin pulldown assays revealed an interaction between IFT20 and Ctnnal1 or Numb. Loss of IFT20 lowered the expression of actin-associated Tagln2, whose knockdown promoted cell migration. Thus, the extraciliary function of ITF20 in breast cancer cell was associated with the negative regulation of migration.
Collapse
Affiliation(s)
- Huihui Yang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fan Zhang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huan Long
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Yiwen Lin
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiahui Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education (Hubei-MOST & KLOBM), Wuhan University, Wuhan, China
| | - Haibin Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education (Hubei-MOST & KLOBM), Wuhan University, Wuhan, China
| | - Kaiyao Huang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
9
|
Rajala A, Rajala RVS. A non-canonical rhodopsin-mediated insulin receptor signaling pathway in retinal photoreceptor neurons. Cell Biol Int 2020; 44:1020-1027. [PMID: 31889373 DOI: 10.1002/cbin.11299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/28/2019] [Indexed: 01/08/2023]
Abstract
We previously reported a ligand-independent and rhodopsin-dependent insulin receptor (IR) neuroprotective signaling pathway in both rod and cone photoreceptor cells, which is activated through protein-protein interaction. Our previous studies were performed with either retina or isolated rod or cone outer segment preparations and the expression of IR signaling proteins were examined. The isolation of outer segments with large portions of the attached inner segments is a technical challenge. Optiprep™ density gradient medium has been used to isolate the cells and subcellular organelles, Optiprep™ is a non-ionic iodixanol-based medium with a density of 1.320 g/mL. We employed this method to examine the expression of IR and its signaling proteins, and activation of one of the downstream effectors of the IR in isolated photoreceptor cells. Identification of the signaling complexes will be helpful for therapeutic targeting in disease conditions.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK, 73104, USA
| | - Raju V S Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma, OK, 73104, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma, OK, 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK, 73104, USA
| |
Collapse
|
10
|
Listik E, Xavier EG, Silva Pinhal MAD, Toma L. Dermatan sulfate epimerase 1 expression and mislocalization may interfere with dermatan sulfate synthesis and breast cancer cell growth. Carbohydr Res 2020; 488:107906. [PMID: 31972438 DOI: 10.1016/j.carres.2020.107906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/10/2019] [Accepted: 01/03/2020] [Indexed: 11/18/2022]
Abstract
Dermatan sulfate (DS) is a glycosaminoglycan (GAG) that is produced through the epimerization of the glucuronic acid on chondroitin sulfate into iduronic acid (IduA) by dermatan sulfate epimerase (DS-epi) 1 and 2. Proteoglycans (PGs) play essential physiological and pathological roles during cellular development, proliferation, differentiation, and cancer metastasis. DS proteoglycans play vital roles during the process of tumorigenesis, due to the increased flexibility of the polysaccharide chain in the presence of IduA residues, which facilitate specific interactions with proteins, such as growth factors, cytokines, and angiogenic factors. Furthermore, DS-epi is highly expressed in many tumors, especially in esophageal squamous cell carcinoma. This study aimed to investigate the expression of DS-epi1 in multiple breast cancer cell lines, including MCF7 (luminal A), MDA-MB-231 (triple-negative) and SKBR3 (human epidermal growth factor receptor 2-positive), and its involvement in cancer progression. A SKBR3 variant, SKBR3m, presented the most erratic cell growth pattern when compared with those for MCF7 and MDA-MB-231. Moreover, SKBR3m cells demonstrated the highest level of DS-epi1 gene expression and higher 35S-DS content. However, at the protein level, MCF7 cells displayed the highest protein level for DS-epi1, whereas MDA-MB-231 cells had the lowest level. DS-epi1 was found in vesicles and in the perinuclear compartment only in SKBR3m cells, suggesting localization in the Golgi apparatus in these cells, in contrast with the cytoplasmic localization observed in MCF7 and MDA-MB-231 cells. The cytoplasm location of DS-epi1 likely compromised the formation of DS chains, but the core protein was detected using a decorin antibody. Golgi-specific labeling confirmed the localization of DS-epi1 in SKBR3m cells at the Golgi apparatus, indicating that the location of the enzyme was a determinant for the synthesis of DS in this cell line, suggesting that DS may play a decisive role in the tumor growth observed in this breast cancer cell line.
Collapse
Affiliation(s)
- Eduardo Listik
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, São Paulo, SP, Brazil, CEP: 04044-020.
| | - Everton Galvão Xavier
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, São Paulo, SP, Brazil, CEP: 04044-020.
| | - Maria Aparecida da Silva Pinhal
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, São Paulo, SP, Brazil, CEP: 04044-020.
| | - Leny Toma
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, São Paulo, SP, Brazil, CEP: 04044-020.
| |
Collapse
|
11
|
Pothukuchi P, Agliarulo I, Russo D, Rizzo R, Russo F, Parashuraman S. Translation of genome to glycome: role of the Golgi apparatus. FEBS Lett 2019; 593:2390-2411. [PMID: 31330561 DOI: 10.1002/1873-3468.13541] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022]
Abstract
Glycans are one of the four biopolymers of the cell and they play important roles in cellular and organismal physiology. They consist of both linear and branched structures and are synthesized in a nontemplated manner in the secretory pathway of mammalian cells with the Golgi apparatus playing a key role in the process. In spite of the absence of a template, the glycans synthesized by a cell are not a random collection of possible glycan structures but a distribution of specific glycans in defined quantities that is unique to each cell type (Cell type here refers to distinct cell forms present in an organism that can be distinguished based on morphological, phenotypic and/or molecular criteria.) While information to produce cell type-specific glycans is encoded in the genome, how this information is translated into cell type-specific glycome (Glycome refers to the quantitative distribution of all glycan structures present in a given cell type.) is not completely understood. We summarize here the factors that are known to influence the fidelity of glycan biosynthesis and integrate them into known glycosylation pathways so as to rationalize the translation of genetic information to cell type-specific glycome.
Collapse
Affiliation(s)
- Prathyush Pothukuchi
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| | - Ilenia Agliarulo
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| | - Domenico Russo
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| | - Riccardo Rizzo
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| | - Francesco Russo
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| | - Seetharaman Parashuraman
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Napoli, Italy
| |
Collapse
|
12
|
Pantazopoulou A, Glick BS. A Kinetic View of Membrane Traffic Pathways Can Transcend the Classical View of Golgi Compartments. Front Cell Dev Biol 2019; 7:153. [PMID: 31448274 PMCID: PMC6691344 DOI: 10.3389/fcell.2019.00153] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/22/2019] [Indexed: 01/07/2023] Open
Abstract
A long-standing assumption is that the cisternae of the Golgi apparatus can be grouped into functionally distinct compartments, yet the molecular identities of those compartments have not been clearly described. The concept of a compartmentalized Golgi is challenged by the cisternal maturation model, which postulates that cisternae form de novo and then undergo progressive biochemical changes. Cisternal maturation can potentially be reconciled with Golgi compartmentation by defining compartments as discrete kinetic stages in the maturation process. These kinetic stages are distinguished by the traffic pathways that are operating. For example, a major transition occurs when a cisterna stops producing COPI vesicles and begins producing clathrin-coated vesicles. This transition separates one kinetic stage, the "early Golgi," from a subsequent kinetic stage, the "late Golgi" or "trans-Golgi network (TGN)." But multiple traffic pathways drive Golgi maturation, and the periods of operation for different traffic pathways can partially overlap, so there is no simple way to define a full set of Golgi compartments in terms of kinetic stages. Instead, we propose that the focus should be on the series of transitions experienced by a Golgi cisterna as various traffic pathways are switched on and off. These traffic pathways drive changes in resident transmembrane protein composition. Transitions in traffic pathways seem to be the fundamental, conserved determinants of Golgi organization. According to this view, the initial goal is to identify the relevant traffic pathways and place them on the kinetic map of Golgi maturation, and the ultimate goal is to elucidate the logic circuit that switches individual traffic pathways on and off as a cisterna matures.
Collapse
Affiliation(s)
- Areti Pantazopoulou
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, United States
| | - Benjamin S Glick
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Mani M, Thao DT, Kim BC, Lee UH, Kim DJ, Jang SH, Back SH, Lee BJ, Cho WJ, Han IS, Park JW. DRG2 knockdown induces Golgi fragmentation via GSK3β phosphorylation and microtubule stabilization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1463-1474. [PMID: 31199931 DOI: 10.1016/j.bbamcr.2019.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/19/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023]
Abstract
The perinuclear stacks of the Golgi apparatus maintained by dynamic microtubules are essential for cell migration. Activation of Akt (protein kinase B, PKB) negatively regulates glycogen synthase kinase 3β (GSK3β)-mediated tau phosphorylation, which enhances tau binding to microtubules and microtubule stability. In this study, experiments were performed on developmentally regulated GTP-binding protein 2 (DRG2)-stably knockdown HeLa cells to determine whether knockdown of DRG2 in HeLa cells treated with epidermal growth factor (EGF) affects microtubule dynamics, perinuclear Golgi stacking, and cell migration. Here, we show that DRG2 plays a key role in regulating microtubule stability, perinuclear Golgi stack formation, and cell migration. DRG2 knockdown prolonged the EGF receptor (EGFR) localization in endosome, enhanced Akt activity and inhibitory phosphorylation of GSK3β. Tau, a target of GSK3β, was hypo-phosphorylated in DRG2-knockdown cells and showed greater association with microtubules, resulting in microtubule stabilization. DRG2-knockdown cells showed defects in microtubule growth and microtubule organizing centers (MTOC), Golgi fragmentation, and loss of directional cell migration. These results reveal a previously unappreciated role for DRG2 in the regulation of perinuclear Golgi stacking and cell migration via its effects on GSK3β phosphorylation, and microtubule stability.
Collapse
Affiliation(s)
- Muralidharan Mani
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Dang Thi Thao
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Beom Chang Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Dong Jun Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Soo Hwa Jang
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Sung Hoon Back
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Wha Ja Cho
- Metainflammation Research Center, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - In-Seob Han
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea.
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea.
| |
Collapse
|
14
|
Kellokumpu S. Golgi pH, Ion and Redox Homeostasis: How Much Do They Really Matter? Front Cell Dev Biol 2019; 7:93. [PMID: 31263697 PMCID: PMC6584808 DOI: 10.3389/fcell.2019.00093] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Exocytic and endocytic compartments each have their own unique luminal ion and pH environment that is important for their normal functioning. A failure to maintain this environment - the loss of homeostasis - is not uncommon. In the worst case, all the main Golgi functions, including glycosylation, membrane trafficking and protein sorting, can be perturbed. Several factors contribute to Golgi homeostasis. These include not only ions such as H+, Ca2+, Mg2+, Mn2+, but also Golgi redox state and nitric oxide (NO) levels, both of which are dependent on the oxygen levels in the cells. Changes to any one of these factors have consequences on Golgi functions, the nature of which can be dissimilar or similar depending upon the defects themselves. For example, altered Golgi pH homeostasis gives rise to Cutis laxa disease, in which glycosylation and membrane trafficking are both affected, while altered Ca2+ homeostasis due to the mutated SCPA1 gene in Hailey-Hailey disease, perturbs various protein sorting, proteolytic cleavage and membrane trafficking events in the Golgi. This review gives an overview of the molecular machineries involved in the maintenance of Golgi ion, pH and redox homeostasis, followed by a discussion of the organelle dysfunction and disease that frequently result from their breakdown. Congenital disorders of glycosylation (CDGs) are discussed only when they contribute directly to Golgi pH, ion or redox homeostasis. Current evidence emphasizes that, rather than being mere supporting factors, Golgi pH, ion and redox homeostasis are in fact key players that orchestrate and maintain all Golgi functions.
Collapse
Affiliation(s)
- Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
15
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
16
|
Dong R, Saheki Y, Swarup S, Lucast L, Harper JW, De Camilli P. Endosome-ER Contacts Control Actin Nucleation and Retromer Function through VAP-Dependent Regulation of PI4P. Cell 2017; 166:408-423. [PMID: 27419871 DOI: 10.1016/j.cell.2016.06.037] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 04/15/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022]
Abstract
VAP (VAPA and VAPB) is an evolutionarily conserved endoplasmic reticulum (ER)-anchored protein that helps generate tethers between the ER and other membranes through which lipids are exchanged across adjacent bilayers. Here, we report that by regulating PI4P levels on endosomes, VAP affects WASH-dependent actin nucleation on these organelles and the function of the retromer, a protein coat responsible for endosome-to-Golgi traffic. VAP is recruited to retromer budding sites on endosomes via an interaction with the retromer SNX2 subunit. Cells lacking VAP accumulate high levels of PI4P, actin comets, and trans-Golgi proteins on endosomes. Such defects are mimicked by downregulation of OSBP, a VAP interactor and PI4P transporter that participates in VAP-dependent ER-endosomes tethers. These results reveal a role of PI4P in retromer-/WASH-dependent budding from endosomes. Collectively, our data show how the ER can control budding dynamics and association with the cytoskeleton of another membrane by direct contacts leading to bilayer lipid modifications.
Collapse
Affiliation(s)
- Rui Dong
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yasunori Saheki
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sharan Swarup
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Louise Lucast
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA; Kavli Institute for Neurosciences, Yale University School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
17
|
Häupl B, Ihling CH, Sinz A. Protein Interaction Network of Human Protein Kinase D2 Revealed by Chemical Cross-Linking/Mass Spectrometry. J Proteome Res 2016; 15:3686-3699. [PMID: 27559607 DOI: 10.1021/acs.jproteome.6b00513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We investigated the interaction network of human PKD2 in the cytosol and in Golgi-enriched subcellular protein fractions by an affinity enrichment strategy combined with chemical cross-linking/mass spectrometry (MS). Analysis of the subproteomes revealed the presence of distinct proteins in the cytosolic and Golgi fractions. The covalent fixation of transient or weak interactors by chemical cross-linking allowed capturing interaction partners that might otherwise disappear during conventional pull-down experiments. In total, 31 interaction partners were identified for PKD2, including glycogen synthase kinase-3 beta (GSK3B), 14-3-3 protein gamma (YWHAG), and the alpha isoform of 55 kDa regulatory subunit B of protein phosphatase 2A (PPP2R2A). Remarkably, the entire seven-subunit Arp2/3 complex (ARPC1B, ARPC2, ARPC3, ARPC4, ARPC5, ACTR3, ACTR2) as well as ARPC1A and ARPC5L, which are putative substitutes of ARPC1B and ARPC5, were identified. We provide evidence of a direct protein-protein interaction between PKD2 and Arp2/3. Our findings will pave the way for further structural and functional studies of PKD2 complexes, especially the PKD2/Arp2/3 interaction, to elucidate the role of PKD2 for transport processes at the trans-Golgi network. Data are available via ProteomeXchange with identifiers PXD003909 (enrichment from cytosolic fractions), PXD003913 (enrichment from Golgi fractions), and PXD003917 (subcellular fractionation).
Collapse
Affiliation(s)
- Björn Häupl
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4, D-06120 Halle (Saale), Germany
| | - Christian H Ihling
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4, D-06120 Halle (Saale), Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4, D-06120 Halle (Saale), Germany
| |
Collapse
|
18
|
Carlson K, Pomerantz SC, Vafa O, Naso M, Strohl W, Mains RE, Eipper BA. Optimizing production of Fc-amidated peptides by Chinese hamster ovary cells. BMC Biotechnol 2015; 15:95. [PMID: 26475607 PMCID: PMC4609047 DOI: 10.1186/s12896-015-0210-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/01/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amidation of the carboxyl terminal of many peptides is essential for full biological potency, often increasing receptor binding and stability. The single enzyme responsible for this reaction is peptidylglycine α-amidating monooxygenase (PAM: EC 1.14.17.3), a copper- and ascorbate-dependent Type I membrane protein. METHODS To make large amounts of high molecular weight amidated product, Chinese hamster ovary (CHO) cells were engineered to express exogenous PAM. To vary access of the enzyme to its substrate, exogenous PAM was targeted to the endoplasmic reticulum, trans-Golgi network, endosomes and lysosomes or to the lumen of the secretory pathway. RESULTS PAM was equally active when targeted to each intracellular location and assayed in homogenates. Immunocytochemical analyses of CHO cells and a pituitary cell line demonstrated that targeting of exogenous PAM was partially successful. PAM substrates generated by expressing peptidylglycine substrates (glucagon-like peptide 1-Gly, peptide YY-Gly and neuromedin U-Gly) fused to the C-terminus of immunoglobulin Fc in CHO cell lines producing targeted PAM. The extent of amidation of the Fc-peptides was determined by mass spectrometry and amidation-specific enzyme immunoassays. Amidation was inhibited by copper chelation, but was not enhanced by the addition of additional copper or ascorbate. CONCLUSIONS Peptide amidation was increased over endogenous levels by exogenous PAM, and targeting PAM to the endoplasmic reticulum or trans-Golgi network increased peptide amidation compared to endogenous CHO PAM.
Collapse
Affiliation(s)
- Kristina Carlson
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| | - Steven C Pomerantz
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Omid Vafa
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Michael Naso
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - William Strohl
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA. .,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
19
|
Kato S, Chen J, Cornog KH, Zhang H, Roberts JD. The Golgi apparatus regulates cGMP-dependent protein kinase I compartmentation and proteolysis. Am J Physiol Cell Physiol 2015; 308:C944-58. [PMID: 25855081 DOI: 10.1152/ajpcell.00199.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 03/31/2015] [Indexed: 01/12/2023]
Abstract
cGMP-dependent protein kinase I (PKGI) is an important effector of cGMP signaling that regulates vascular smooth muscle cell (SMC) phenotype and proliferation. PKGI has been detected in the perinuclear region of cells, and recent data indicate that proprotein convertases (PCs) typically resident in the Golgi apparatus (GA) can stimulate PKGI proteolysis and generate a kinase fragment that localizes to the nucleus and regulates gene expression. However, the role of the endomembrane system in PKGI compartmentation and processing is unknown. Here, we demonstrate that PKGI colocalizes with endoplasmic reticulum (ER), ER-Golgi intermediate compartment, GA cisterna, and trans-Golgi network proteins in pulmonary artery SMC and cell lines. Moreover, PKGI localizes with furin, a trans-Golgi network-resident PC known to cleave PKGI. ER protein transport influences PKGI localization because overexpression of a constitutively inactive Sar1 transgene caused PKGI retention in the ER. Additionally, PKGI appears to reside within the GA because PKGI immunoreactivity was determined to be resistant to cytosolic proteinase K treatment in live cells. The GA appears to play a role in PKGI proteolysis because overexpression of inositol 1,4,5-trisphosphate receptor-associated cGMP kinase substrate, not only tethered heterologous PKGI-β to the ER and decreased its localization to the GA, but also diminished PKGI proteolysis and nuclear translocation. Also, inhibiting intra-GA protein transport with monensin was observed to decrease PKGI cleavage. These studies detail a role for the endomembrane system in regulating PKGI compartmentation and proteolysis. Moreover, they support the investigation of mechanisms regulating PKGI-dependent nuclear cGMP signaling in the pulmonary vasculature with Golgi dysfunction.
Collapse
Affiliation(s)
- Shin Kato
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Jingsi Chen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Huili Zhang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Jesse D Roberts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts; Departments of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Cambridge, Massachusetts;
| |
Collapse
|
20
|
Isaji T, Im S, Gu W, Wang Y, Hang Q, Lu J, Fukuda T, Hashii N, Takakura D, Kawasaki N, Miyoshi H, Gu J. An oncogenic protein Golgi phosphoprotein 3 up-regulates cell migration via sialylation. J Biol Chem 2015; 289:20694-705. [PMID: 24895123 DOI: 10.1074/jbc.m113.542688] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently, the Golgi phosphoprotein 3 (GOLPH3) and its yeast homolog Vps74p have been characterized as essential for the Golgi localization of glycosyltransferase in yeast. GOLPH3 has been identified as a new oncogene that is commonly amplified in human cancers to modulate mammalian target of rapamycin signaling. However, the molecular mechanisms of the carcinogenic signaling pathway remain largely unclear. To investigate whether the expression of GOLPH3 was involved in the glycosylation processes in mammalian cells, and whether it affected cell behavior, we performed a loss-of-function study. Cell migration was suppressed in GOLPH3 knockdown (KD) cells, and the suppression was restored by a re-introduction of the GOLPH3 gene. HPLC and LC/MS analysis showed that the sialylation of N-glycans was specifically decreased in KD cells. The specific interaction between sialyltransferases and GOLPH3 was important for the sialylation. Furthermore, overexpression of α2,6-sialyltransferase-I rescued cell migration and cellular signaling, both of which were blocked in GOLPH3 knockdown cells. These results are the first direct demonstration of the role of GOLPH3 in N-glycosylation to regulate cell biological functions.
Collapse
|
21
|
Eckert ESP, Reckmann I, Hellwig A, Röhling S, El-Battari A, Wieland FT, Popoff V. Golgi phosphoprotein 3 triggers signal-mediated incorporation of glycosyltransferases into coatomer-coated (COPI) vesicles. J Biol Chem 2014; 289:31319-29. [PMID: 25246532 PMCID: PMC4223332 DOI: 10.1074/jbc.m114.608182] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Indexed: 12/30/2022] Open
Abstract
Newly synthesized membrane and secreted proteins undergo a series of posttranslational modifications in the Golgi apparatus, including attachment of carbohydrate moieties. The final structure of so-formed glycans is determined by the order of execution of the different glycosylation steps, which seems intimately related to the spatial distribution of glycosyltransferases and glycosyl hydrolases within the Golgi apparatus. How cells achieve an accurate localization of these enzymes is not completely understood but might involve dynamic processes such as coatomer-coated (COPI) vesicle-mediated trafficking. In yeast, this transport is likely to be regulated by vacuolar protein sorting 74 (Vps74p), a peripheral Golgi protein able to interact with COPI coat as well as with a binding motif present in the cytosolic tails of some mannosyltransferases. Recently, Golgi phosphoprotein 3 (GOLPH3), the mammalian homolog of Vps74, has been shown to control the Golgi localization of core 2 N-acetylglucosamine-transferase 1. Here, we highlight a role of GOLPH3 in the spatial localization of α-2,6-sialyltransferase 1. We show, for the first time, that GOLPH3 supports incorporation of both core 2 N-acetylglucosamine-transferase 1 and α-2,6-sialyltransferase 1 into COPI vesicles. Depletion of GOLPH3 altered the subcellular localization of these enzymes. In contrast, galactosyltransferase, an enzyme that does not interact with GOLPH3, was neither incorporated into COPI vesicles nor was dependent on GOLPH3 for proper localization.
Collapse
Affiliation(s)
- Elias S P Eckert
- From the Heidelberg University Biochemistry Center (BZH), INF 328 and
| | - Ingeborg Reckmann
- From the Heidelberg University Biochemistry Center (BZH), INF 328 and
| | - Andrea Hellwig
- Interdisciplinary Center for Neurosciences (IZN), INF 364, Heidelberg University, 69120 Heidelberg, Germany and
| | - Simone Röhling
- From the Heidelberg University Biochemistry Center (BZH), INF 328 and
| | - Assou El-Battari
- INSERM UMR 911, Aix-Marseille Université, Centre de Recherche en Oncobiologie et Oncopharmacologie (CR02), 13284 Marseille, France
| | - Felix T Wieland
- From the Heidelberg University Biochemistry Center (BZH), INF 328 and
| | - Vincent Popoff
- From the Heidelberg University Biochemistry Center (BZH), INF 328 and
| |
Collapse
|
22
|
Romero AM, Renau-Piqueras J, Marín MP, Esteban-Pretel G. Chronic alcohol exposure affects the cell components involved in membrane traffic in neuronal dendrites. Neurotox Res 2014; 27:43-54. [PMID: 25022897 DOI: 10.1007/s12640-014-9484-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/13/2014] [Accepted: 07/01/2014] [Indexed: 01/09/2023]
Abstract
The specific traffic of the membrane components in neurons is a major requirement to establish and maintain neuronal domains-the axonal and the somatodendritic domains-and their polarized morphology. Unlike axons, dendrites contain membranous organelles, which are involved in the secretory pathway, including the endoplasmic reticulum, the Golgi apparatus and post-Golgi apparatus carriers, the cytoskeleton, and plasma membrane. A variety of molecules and factors are also involved in this process. Previous studies have shown that chronic alcohol exposure negatively affects several of these cell components, such as the Golgi apparatus or cytoskeleton in neurons. Yet very little information is available on the possible effects of this exposure on the remaining cell elements involved in intracellular trafficking in neurons, particularly in dendrites. By qualitative and quantitative electron microscopy, immunofluorescence and immunoblotting, we herein show that chronic exposure to moderate levels (30 mM) of ethanol in cultured neurons reduces the volume and surface density of the rough endoplasmic reticulum, and increases the levels of GRP78, a chaperone involved in endoplasmic reticulum stress. Ethanol also significantly diminishes the proportion of neurons that show an extension of Golgi into dendrites and dendritic Golgi outposts, a structure present exclusively in longer, thicker apical dendrites. Both Golgi apparatus types were also fragmented into a large number of cells. We also investigated the effect of alcohol on the levels of microtubule-based motor proteins KIF5, KIF17, KIFC2, dynein, and myosin IIb, responsible for transporting different cargoes in dendrites. Of these, alcohol differently affects several of them by lowering dynein and raising KIF5, KIFC2, and myosin IIb. These results, together with other previously published ones, suggest that practically all the protein trafficking steps in dendrites are altered to a greater or lesser extent by chronic alcohol exposure in neuronal cells, which may have negative repercussions for the development and maintenance of their polarized morphology and function.
Collapse
Affiliation(s)
- Ana M Romero
- Sección de Biología y Patología Celular, Centro de Investigación, Hospital Universitario ''La Fe'', Avenida Campanar 21, 46009, Valencia, Spain
| | | | | | | |
Collapse
|
23
|
Ong YS, Tran THT, Gounko NV, Hong W. TMEM115 is an integral membrane protein of the Golgi complex involved in retrograde transport. J Cell Sci 2014; 127:2825-39. [PMID: 24806965 PMCID: PMC4077589 DOI: 10.1242/jcs.136754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Searching and evaluating the Human Protein Atlas for transmembrane proteins enabled us to identify an integral membrane protein, TMEM115, that is enriched in the Golgi complex. Biochemical and cell biological analysis suggested that TMEM115 has four candidate transmembrane domains located in the N-terminal region. Both the N- and C-terminal domains are oriented towards the cytoplasm. Immunofluorescence analysis supports that TMEM115 is enriched in the Golgi cisternae. Functionally, TMEM115 knockdown or overexpression delays Brefeldin-A-induced Golgi-to-ER retrograde transport, phenocopying cells with mutations or silencing of the conserved oligomeric Golgi (COG) complex. Co-immunoprecipitation and in vitro binding experiments reveals that TMEM115 interacts with the COG complex, and might self-interact to form dimers or oligomers. A short region (residues 206–229) immediately to the C-terminal side of the fourth transmembrane domain is both necessary and sufficient for Golgi targeting. Knockdown of TMEM115 also reduces the binding of the lectins peanut agglutinin (PNA) and Helix pomatia agglutinin (HPA), suggesting an altered O-linked glycosylation profile. These results establish that TMEM115 is an integral membrane protein of the Golgi stack regulating Golgi-to-ER retrograde transport and is likely to be part of the machinery of the COG complex.
Collapse
Affiliation(s)
- Yan Shan Ong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Ton Hoai Thi Tran
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore IMB-IMCB Joint Electron Microscopy Suite, 20 Biopolis Street, Singapore 138671, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore Department of Biochemistry, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
24
|
Pellett PA, Dietrich F, Bewersdorf J, Rothman JE, Lavieu G. Inter-Golgi transport mediated by COPI-containing vesicles carrying small cargoes. eLife 2013; 2:e01296. [PMID: 24137546 PMCID: PMC3787390 DOI: 10.7554/elife.01296] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 08/21/2013] [Indexed: 01/15/2023] Open
Abstract
A core prediction of the vesicular transport model is that COPI vesicles are responsible for trafficking anterograde cargoes forward. In this study, we test this prediction by examining the properties and requirements of inter-Golgi transport within fused cells, which requires mobile carriers in order for exchange of constituents to occur. We report that both small soluble and membrane-bound secretory cargo and exogenous Golgi resident glycosyl-transferases are exchanged between separated Golgi. Large soluble aggregates, which traverse individual stacks, do not transfer between Golgi, implying that small cargoes (which can fit in a typical transport vesicle) are transported by a different mechanism. Super-resolution microscopy reveals that the carriers of both anterograde and retrograde cargoes are the size of COPI vesicles, contain coatomer, and functionally require ARF1 and coatomer for transport. The data suggest that COPI vesicles traffic both small secretory cargo and steady-state Golgi resident enzymes among stacked cisternae that are stationary. DOI:http://dx.doi.org/10.7554/eLife.01296.001 All eukaryotic cells contain an organelle called the Golgi apparatus, which consists of a series of four to six flattened structures called cisternae. Proteins that are intended for secretion from the cell, or proteins that go on to become part of the cell membrane, must pass through the Golgi, where they undergo modifications that ensure they are targeted to the correct place. There are two main models for how proteins are transported from the entry side of the Golgi, known as the cis face, to the exit side (trans face), through a process known as anterograde transport. One possibility is that the cargo protein matures within a single cisterna, which gradually moves from the cis to the trans face without the protein ever leaving it. Alternatively, the cisternae may remain fixed in position, while individual proteins are carried between them by specialized transport vesicles called COPI vesicles. Now, Pellett et al. have used modern molecular biology techniques to revisit this question, more than 25 years after members of the same group first obtained evidence suggesting the involvement of COPI vesicles. To do this, they labelled the proteins that reside within the Golgi of one cell green, and those within the Golgi of another cell, red. They then fused the two cells together, and traced the movement of labelled proteins between the two organelles. Proteins that are known to undergo anterograde transport were also transported between the two Golgi, whereas large protein aggregates were not. Super-resolution microscopy revealed that the transported proteins were carried in vesicles the size of COPI vesicles and surrounded by a coat protein that resembles COPI. Moreover, transport involved the adaptor protein ARF, which helps to load cargo into COPI vesicles. By providing evidence that Golgi resident proteins and proteins that normally undergo anterograde transport can be carried by COPI vesicles between two physically separate Golgi, Pellett et al. increase the weight of evidence that COPI vesicles may also be responsible for both retrograde and anterograde transport within the Golgi itself. DOI:http://dx.doi.org/10.7554/eLife.01296.002
Collapse
Affiliation(s)
- Patrina A Pellett
- Department of Cell Biology , Yale University School of Medicine , New Haven , United States ; Department of Chemistry , Yale University , New Haven , United States
| | | | | | | | | |
Collapse
|
25
|
Petrosyan A, Cheng PW. A non-enzymatic function of Golgi glycosyltransferases: mediation of Golgi fragmentation by interaction with non-muscle myosin IIA. Glycobiology 2013; 23:690-708. [PMID: 23396488 DOI: 10.1093/glycob/cwt009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The Golgi apparatus undergoes morphological changes under stress or malignant transformation, but the precise mechanisms are not known. We recently showed that non-muscle myosin IIA (NMIIA) binds to the cytoplasmic tail of Core 2 N-acetylglucosaminyltransferase mucus-type (C2GnT-M) and transports it to the endoplasmic reticulum for recycling. Here, we report that Golgi fragmentation induced by brefeldin A (BFA) or coatomer protein (β-COP) knockdown (KD) in Panc1-bC2GnT-M (c-Myc) cells is accompanied by the increased association of NMIIA with C2GnT-M and its degradation by proteasomes. Golgi fragmentation is prevented by inhibition or KD of NMIIA. Using multiple approaches, we have shown that the speed of BFA-induced Golgi fragmentation is positively correlated with the levels of this enzyme in the Golgi. The observation is reproduced in LNCaP cells which express high levels of two endogenous glycosyltransferases--C2GnT-L and β-galactoside α2,3 sialyltransferase 1. NMIIA is found to form complexes with these two enzymes but not Golgi matrix proteins. The KD of both enzymes or the prevention of Golgi glycosyltransferases from exiting endoplasmic reticulum reduced Golgi-associated NMIIA and decreased the BFA-induced fragmentation. Interestingly, the fragmented Golgi detected in colon cancer HT-29 cells can be restored to a compact morphology after inhibition or KD of NMIIA. The Golgi disorganization induced by the microtubule or actin destructive agent is NMIIA-independent and does not affect the levels of glycosyltransferases. We conclude that NMIIA interacts with Golgi residential but not matrix proteins, and this interaction is responsible for Golgi fragmentation induced by β-COP KD or BFA treatment. This is a novel non-enzymatic function of Golgi glycosyltransferases.
Collapse
Affiliation(s)
- Armen Petrosyan
- Department of Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105 USA
| | | |
Collapse
|
26
|
Tien WS, Chen YT, Wu KP. SecretePipe: A Screening Pipeline for Secreted Proteins with Competence to Identify Potential Membrane-Bound Shed Markers. J Proteome Res 2013; 12:1235-44. [PMID: 23336694 DOI: 10.1021/pr3009012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei 112, Taiwan
- Bioinformatics Program, Taiwan International Graduate
Program, Academia Sinica, Taipei 115, Taiwan
| | - Yen-Tsuen Chen
- Institute of Biomedical Informatics, National Yang Ming University, Taipei 112, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei 112, Taiwan
| |
Collapse
|
27
|
Esteban-Pretel G, Marín MP, Romero AM, Timoneda J, Ponsoda X, Ballestín R, Renau-Piqueras J. Polyphosphoinositide metabolism and Golgi complex morphology in hippocampal neurons in primary culture is altered by chronic ethanol exposure. Alcohol Alcohol 2012; 48:15-27. [PMID: 23118092 DOI: 10.1093/alcalc/ags117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
AIMS Ethanol affects not only the cytoskeletal organization and activity, but also intracellular trafficking in neurons in the primary culture. Polyphosphoinositide (PPIn) are essential regulators of many important cell functions, including those mentioned, cytoskeleton integrity and intracellular vesicle trafficking. Since information about the effect of chronic ethanol exposure on PPIn metabolism in neurons is scarce, this study analysed the effect of this treatment on three of these phospholipids. METHODS Phosphatidylinositol (PtdIns) levels as well as the activity and/or levels of enzymes involved in their metabolism were analysed in neurons chronically exposed to ethanol. The levels of phospholipases C and D, and phosphatidylethanol formation were also assessed. The consequence of the possible alterations in the levels of PtdIns on the Golgi complex (GC) was also analysed. RESULTS We show that phosphatidylinositol (4,5)-bisphosphate and phosphatidylinositol (3,4,5)-trisphosphate levels, both involved in the control of intracellular trafficking and cytoskeleton organization, decrease in ethanol-exposed hippocampal neurons. In contrast, several kinases that participate in the metabolism of these phospholipids, and the level and/or activity of phospholipases C and D, increase in cells after ethanol exposure. Ethanol also promotes phosphatidylethanol formation in neurons, which can result in the suppression of phosphatidic acid synthesis and, therefore, in PPIn biosynthesis. This treatment also lowers the phosphatidylinositol 4-phosphate levels, the main PPIn in the GC, with alterations in their morphology and in the levels of some of the proteins involved in structure maintenance. CONCLUSIONS The deregulation of the metabolism of PtdIns may underlie the ethanol-induced alterations on different neuronal processes, including intracellular trafficking and cytoskeletal integrity.
Collapse
Affiliation(s)
- Guillermo Esteban-Pretel
- Corresponding author: Sección de Biología y Patología Celular, Centro de Investigación, Hospital Universitario La Fe, Avda. Campanar 21, Valencia, Spain.
| | | | | | | | | | | | | |
Collapse
|
28
|
Petrosyan A, Ali MF, Verma SK, Cheng H, Cheng PW. Non-muscle myosin IIA transports a Golgi glycosyltransferase to the endoplasmic reticulum by binding to its cytoplasmic tail. Int J Biochem Cell Biol 2012; 44:1153-65. [PMID: 22525330 PMCID: PMC4011501 DOI: 10.1016/j.biocel.2012.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/14/2012] [Accepted: 04/04/2012] [Indexed: 12/18/2022]
Abstract
The mechanism of the Golgi-to-ER transport of Golgi glycosyltransferases is not clear. We utilize a cell line expressing the core 2 N-acetylglucosaminyltransferase-M (C2GnT-M) tagged with c-Myc to explore this mechanism. By immunoprecipitation using anti-c-Myc antibodies coupled with proteomics analysis, we have identified several proteins including non-muscle myosin IIA (NMIIA), heat shock protein (HSP)-70 and ubiquitin activating enzyme E1 in the immunoprecipitate. Employing yeast-two-hybrid analysis and pulldown experiments, we show that the C-terminal region of the NMIIA heavy chain binds to the 1-6 amino acids in the cytoplasmic tail of C2GnT-M. We have found that NMIIA co-localizes with C2GnT-M at the periphery of the Golgi. In addition, inhibition or knockdown of NMIIA prevents the brefeldin A-induced collapse of the Golgi as shown by the inhibition of the migration of both Giantin, a Golgi matrix protein, and C2GnT-M, a Golgi non-matrix protein, to the ER. In contrast, knockdown of HSP70 retains Giantin in the Golgi but moves C2GnT-M to the ER, a process also blocked by inhibition or knockdown of NMIIA. Also, the intracellular distribution of C2GnT-M is not affected by knockdown of β-coatomer protein with or without inhibition of HSPs, suggesting that the Golgi-to-ER trafficking of C2GnT-M does not depend on coat protein complex-I. Further, inhibition of proteasome results in accumulation of ubiquitinated C2GnT-M, suggesting its degradation by proteasome. Therefore, NMIIA and not coat protein complex-I is responsible for transporting the Golgi glycosyltransferase to the ER for proteasomal degradation. The data suggest that NMIIA is involved in the Golgi remodeling.
Collapse
Affiliation(s)
- Armen Petrosyan
- Omaha Western Iowa Health System, VA Service, Department of Veterans Affairs Medical Center, Omaha, NE, USA
| | | | | | | | | |
Collapse
|
29
|
Srinivasan R, Richards CI, Xiao C, Rhee D, Pantoja R, Dougherty DA, Miwa JM, Lester HA. Pharmacological chaperoning of nicotinic acetylcholine receptors reduces the endoplasmic reticulum stress response. Mol Pharmacol 2012; 81:759-69. [PMID: 22379121 DOI: 10.1124/mol.112.077792] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We report the first observation that endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) can decrease when a central nervous system drug acts as an intracellular pharmacological chaperone for its classic receptor. Transient expression of α4β2 nicotinic receptors (nAChRs) in Neuro-2a cells induced the nuclear translocation of activating transcription factor 6 (ATF6), which is part of the UPR. Cells were exposed for 48 h to the full agonist nicotine, the partial agonist cytisine, or the competitive antagonist dihydro-β-erythroidine; we also tested mutant nAChRs that readily exit the ER. Each of these four manipulations increased Sec24D-enhanced green fluorescent protein fluorescence of condensed ER exit sites and attenuated translocation of ATF6-enhanced green fluorescent protein to the nucleus. However, we found no correlation among the manipulations regarding other tested parameters [i.e., changes in nAChR stoichiometry (α4(2)β2(3) versus α4(3)β2(2)), changes in ER and trans-Golgi structures, or the degree of nAChR up-regulation at the plasma membrane]. The four manipulations activated 0 to 0.4% of nAChRs, which shows that activation of the nAChR channel did not underlie the reduced ER stress. Nicotine also attenuated endogenously expressed ATF6 translocation and phosphorylation of eukaryotic initiation factor 2α in mouse cortical neurons transfected with α4β2 nAChRs. We conclude that, when nicotine accelerates ER export of α4β2 nAChRs, this suppresses ER stress and the UPR. Suppression of a sustained UPR may explain the apparent neuroprotective effect that causes the inverse correlation between a person's history of tobacco use and susceptibility to developing Parkinson's disease. This suggests a novel mechanism for neuroprotection by nicotine.
Collapse
Affiliation(s)
- Rahul Srinivasan
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The protein composition of the Golgi is intimately linked to its structure and function. As the Golgi serves as the major protein-sorting hub for the secretory pathway, it faces the unique challenge of maintaining its protein composition in the face of constant influx and efflux of transient cargo proteins. Much of our understanding of how proteins are retained in the Golgi has come from studies on glycosylation enzymes, largely because of the compartment-specific distributions these proteins display. From these and other studies of Golgi membrane proteins, we now understand that a variety of retention mechanisms are employed, the majority of which involve the dynamic process of iterative rounds of retrograde and anterograde transport. Such mechanisms rely on protein conformation and amino acid-based sorting signals as well as on properties of transmembrane domains and their relationship with the unique lipid composition of the Golgi.
Collapse
Affiliation(s)
- David K Banfield
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, SAR of China.
| |
Collapse
|
31
|
Mizutani A, Tsunashima H, Nishijima KI, Sasamoto T, Yamada Y, Kojima Y, Motono M, Kojima J, Inayoshi Y, Miyake K, Park EY, Iijima S. Genetic modification of a chicken expression system for the galactosylation of therapeutic proteins produced in egg white. Transgenic Res 2011; 21:63-75. [DOI: 10.1007/s11248-011-9511-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 03/30/2011] [Indexed: 01/12/2023]
|
32
|
Srinivasan R, Pantoja R, Moss FJ, Mackey EDW, Son CD, Miwa J, Lester HA. Nicotine up-regulates alpha4beta2 nicotinic receptors and ER exit sites via stoichiometry-dependent chaperoning. ACTA ACUST UNITED AC 2011; 137:59-79. [PMID: 21187334 PMCID: PMC3010053 DOI: 10.1085/jgp.201010532] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The up-regulation of α4β2* nicotinic acetylcholine receptors (nAChRs) by chronic nicotine is a cell-delimited process and may be necessary and sufficient for the initial events of nicotine dependence. Clinical literature documents an inverse relationship between a person’s history of tobacco use and his or her susceptibility to Parkinson’s disease; this may also result from up-regulation. This study visualizes and quantifies the subcellular mechanisms involved in nicotine-induced nAChR up-regulation by using transfected fluorescent protein (FP)-tagged α4 nAChR subunits and an FP-tagged Sec24D endoplasmic reticulum (ER) exit site marker. Total internal reflection fluorescence microscopy shows that nicotine (0.1 µM for 48 h) up-regulates α4β2 nAChRs at the plasma membrane (PM), despite increasing the fraction of α4β2 nAChRs that remain in near-PM ER. Pixel-resolved normalized Förster resonance energy transfer microscopy between α4-FP subunits shows that nicotine stabilizes the (α4)2(β2)3 stoichiometry before the nAChRs reach the trans-Golgi apparatus. Nicotine also induces the formation of additional ER exit sites (ERES). To aid in the mechanistic analysis of these phenomena, we generated a β2enhanced-ER-export mutant subunit that mimics two regions of the β4 subunit sequence: the presence of an ER export motif and the absence of an ER retention/retrieval motif. The α4β2enhanced-ER-export nAChR resembles nicotine-exposed nAChRs with regard to stoichiometry, intracellular mobility, ERES enhancement, and PM localization. Nicotine produces only small additional PM up-regulation of α4β2enhanced-ER-export receptors. The experimental data are simulated with a model incorporating two mechanisms: (1) nicotine acts as a stabilizing pharmacological chaperone for nascent α4β2 nAChRs in the ER, eventually increasing PM receptors despite a bottleneck(s) in ER export; and (2) removal of the bottleneck (e.g., by expression of the β2enhanced-ER-export subunit) is sufficient to increase PM nAChR numbers, even without nicotine. The data also suggest that pharmacological chaperoning of nAChRs by nicotine can alter the physiology of ER processes.
Collapse
Affiliation(s)
- Rahul Srinivasan
- Division of Biology MC 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Maccioni HJF, Quiroga R, Ferrari ML. Cellular and molecular biology of glycosphingolipid glycosylation. J Neurochem 2011; 117:589-602. [PMID: 21371037 DOI: 10.1111/j.1471-4159.2011.07232.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brain tissue is characterized by its high glycosphingolipid content, particularly those containing sialic acid (gangliosides). As a result of this observation, brain tissue was a focus for studies leading to the characterization of the enzymes participating in ganglioside biosynthesis, and their participation in driving the compositional changes that occur in glycolipid expression during brain development. Later on, this focus shifted to the study of cellular aspects of the synthesis, which lead to the identification of the site of synthesis in the neuronal soma and their axonal transport toward the periphery. In this review article, we will focus in subcellular aspects of the biosynthesis of glycosphingolipid oligosaccharides, particularly the mechanisms underlying the trafficking of glycosphingolipid glycosyltransferases from the endoplasmic reticulum to the Golgi, those that promote their retention in the Golgi and those that participate in their topological organization as part of the complex membrane bound machinery for the synthesis of glycosphingolipids.
Collapse
Affiliation(s)
- Hugo J F Maccioni
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC (UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | | | | |
Collapse
|
34
|
Maccioni HJF, Quiroga R, Spessott W. Organization of the synthesis of glycolipid oligosaccharides in the Golgi complex. FEBS Lett 2011; 585:1691-8. [PMID: 21420403 DOI: 10.1016/j.febslet.2011.03.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/11/2011] [Accepted: 03/14/2011] [Indexed: 11/28/2022]
Abstract
Glycolipids constitute a complex family of amphipathic molecules structurally characterized by a hydrophilic mono- or oligo-saccharide moiety linked to a hydrophobic ceramide moiety. Due to their asymmetric distribution in cell membranes, exposing the saccharide moiety to the extracytoplasmic side of the cell, glycolipids participate in a variety of cell-cell and cell-ligand interactions. Here we summarize aspects of the cell biology of the stepwise synthesis of the saccharide moiety in the Golgi complex of cells from vertebrates. In particular we refer to the participant glycosyltransferases, with emphasis on their trafficking along the secretory pathway, their retention and organization in the Golgi complex membranes and their dependence on the Golgi complex ultra structural organization for proper function.
Collapse
Affiliation(s)
- Hugo J F Maccioni
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC (UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | | | | |
Collapse
|
35
|
Flannery AR, Czibener C, Andrews NW. Palmitoylation-dependent association with CD63 targets the Ca2+ sensor synaptotagmin VII to lysosomes. ACTA ACUST UNITED AC 2010; 191:599-613. [PMID: 21041449 PMCID: PMC3003310 DOI: 10.1083/jcb.201003021] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Posttranslational lipid modifications promote association of Syt VII with the tetraspanin CD63, determining its exit from the Golgi and targeting to lysosomes. Syt VII is a Ca2+ sensor that regulates lysosome exocytosis and plasma membrane repair. Because it lacks motifs that mediate lysosomal targeting, it is unclear how Syt VII traffics to these organelles. In this paper, we show that mutations or inhibitors that abolish palmitoylation disrupt Syt VII targeting to lysosomes, causing its retention in the Golgi complex. In macrophages, Syt VII is translocated simultaneously with the lysosomal tetraspanin CD63 from tubular lysosomes to nascent phagosomes in a Ca2+-dependent process that facilitates particle uptake. Mutations in Syt VII palmitoylation sites block trafficking of Syt VII, but not CD63, to lysosomes and phagosomes, whereas tyrosine replacement in the lysosomal targeting motif of CD63 causes both proteins to accumulate on the plasma membrane. Complexes of CD63 and Syt VII are detected only when Syt VII palmitoylation sites are intact. These findings identify palmitoylation-dependent association with the tetraspanin CD63 as the mechanism by which Syt VII is targeted to lysosomes.
Collapse
Affiliation(s)
- Andrew R Flannery
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | | | | |
Collapse
|
36
|
Reynders E, Foulquier F, Annaert W, Matthijs G. How Golgi glycosylation meets and needs trafficking: the case of the COG complex. Glycobiology 2010; 21:853-63. [PMID: 21112967 DOI: 10.1093/glycob/cwq179] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Protein glycosylation is one of the major biosynthetic functions occurring in the endoplasmic reticulum and Golgi compartments. It requires an amazing number of enzymes, chaperones, lectins and transporters whose actions delicately secure the fidelity of glycan structures. Over the past 30 years, glycobiologists hammered that glycan structures are not mere decorative elements but serve crucial cellular functions. This becomes dramatically illustrated by a group of mostly severe, inherited human disorders named congenital disorders of glycosylation (CDG). To date, many types of CDG have been defined genetically and most of the time the defects impair the biosynthesis, transfer and remodeling of N-glycans. Recently, the identification of the several types of CDG caused by deficiencies in the conserved oligomeric Golgi (COG) complex, a complex involved in vesicular Golgi trafficking, expanded the field of CDG but also brought novel insights in glycosylation. The molecular mechanisms underlying the complex pathway of N-glycosylation in the Golgi are far from understood. The availability of COG-deficient CDG patients and patients' cells offered a new way to study how COG, and its different subunits, could influence the Golgi N-glycosylation machinery and localization. This review summarizes the recent findings on the implication of COG in Golgi glycosylation. It highlights the need for a dynamic, finely tuned balance between anterograde and retrograde trafficking for the correct localization of Golgi enzymes to assure the stepwise maturation of N-glycan chains.
Collapse
Affiliation(s)
- Ellen Reynders
- Laboratory for Membrane Trafficking, Center for Human Genetics, KULeuven, Department for Molecular and Developmental Genetics (VIB), Leuven, Belgium
| | | | | | | |
Collapse
|
37
|
Schmitz MHA, Held M, Janssens V, Hutchins JRA, Hudecz O, Ivanova E, Goris J, Trinkle-Mulcahy L, Lamond AI, Poser I, Hyman AA, Mechtler K, Peters JM, Gerlich DW. Live-cell imaging RNAi screen identifies PP2A-B55alpha and importin-beta1 as key mitotic exit regulators in human cells. Nat Cell Biol 2010; 12:886-93. [PMID: 20711181 PMCID: PMC3839080 DOI: 10.1038/ncb2092] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 07/02/2010] [Indexed: 01/20/2023]
Abstract
When vertebrate cells exit mitosis various cellular structures are re-organized to build functional interphase cells. This depends on Cdk1 (cyclin dependent kinase 1) inactivation and subsequent dephosphorylation of its substrates. Members of the protein phosphatase 1 and 2A (PP1 and PP2A) families can dephosphorylate Cdk1 substrates in biochemical extracts during mitotic exit, but how this relates to postmitotic reassembly of interphase structures in intact cells is not known. Here, we use a live-cell imaging assay and RNAi knockdown to screen a genome-wide library of protein phosphatases for mitotic exit functions in human cells. We identify a trimeric PP2A-B55alpha complex as a key factor in mitotic spindle breakdown and postmitotic reassembly of the nuclear envelope, Golgi apparatus and decondensed chromatin. Using a chemically induced mitotic exit assay, we find that PP2A-B55alpha functions downstream of Cdk1 inactivation. PP2A-B55alpha isolated from mitotic cells had reduced phosphatase activity towards the Cdk1 substrate, histone H1, and was hyper-phosphorylated on all subunits. Mitotic PP2A complexes co-purified with the nuclear transport factor importin-beta1, and RNAi depletion of importin-beta1 delayed mitotic exit synergistically with PP2A-B55alpha. This demonstrates that PP2A-B55alpha and importin-beta1 cooperate in the regulation of postmitotic assembly mechanisms in human cells.
Collapse
Affiliation(s)
- Michael H. A. Schmitz
- Institute of Biochemistry, Swiss Federal Institute of Technology Zurich (ETHZ), Schafmattstrasse 18, CH-8093 Zurich, Switzerland
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Michael Held
- Institute of Biochemistry, Swiss Federal Institute of Technology Zurich (ETHZ), Schafmattstrasse 18, CH-8093 Zurich, Switzerland
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Molecular Cell Biology, Faculty of Medicine, KU Leuven, Gasthuisberg O&N1, Herestraat 49 Box 901, B-3000 Leuven, Belgium
| | | | - Otto Hudecz
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Elitsa Ivanova
- Laboratory of Protein Phosphorylation and Proteomics, Department of Molecular Cell Biology, Faculty of Medicine, KU Leuven, Gasthuisberg O&N1, Herestraat 49 Box 901, B-3000 Leuven, Belgium
| | - Jozef Goris
- Laboratory of Protein Phosphorylation and Proteomics, Department of Molecular Cell Biology, Faculty of Medicine, KU Leuven, Gasthuisberg O&N1, Herestraat 49 Box 901, B-3000 Leuven, Belgium
| | - Laura Trinkle-Mulcahy
- Department of Cellular & Molecular Medicine and the Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Angus I. Lamond
- Wellcome Trust Centre for Gene Regulation & Expression, MSI/WTB/JBC Complex, University of Dundee, Dundee, DD1 5EH, UK
| | - Ina Poser
- Max-Planck-Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | - Anthony A. Hyman
- Max-Planck-Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | - Karl Mechtler
- Institute of Molecular Pathology, Dr. Bohr-Gasse 7, 1030 Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Jan-Michael Peters
- Institute of Molecular Pathology, Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Daniel W. Gerlich
- Institute of Biochemistry, Swiss Federal Institute of Technology Zurich (ETHZ), Schafmattstrasse 18, CH-8093 Zurich, Switzerland
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| |
Collapse
|
38
|
CellCognition: time-resolved phenotype annotation in high-throughput live cell imaging. Nat Methods 2010; 7:747-54. [PMID: 20693996 DOI: 10.1038/nmeth.1486] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 07/06/2010] [Indexed: 01/20/2023]
Abstract
Fluorescence time-lapse imaging has become a powerful tool to investigate complex dynamic processes such as cell division or intracellular trafficking. Automated microscopes generate time-resolved imaging data at high throughput, yet tools for quantification of large-scale movie data are largely missing. Here we present CellCognition, a computational framework to annotate complex cellular dynamics. We developed a machine-learning method that combines state-of-the-art classification with hidden Markov modeling for annotation of the progression through morphologically distinct biological states. Incorporation of time information into the annotation scheme was essential to suppress classification noise at state transitions and confusion between different functional states with similar morphology. We demonstrate generic applicability in different assays and perturbation conditions, including a candidate-based RNA interference screen for regulators of mitotic exit in human cells. CellCognition is published as open source software, enabling live-cell imaging-based screening with assays that directly score cellular dynamics.
Collapse
|
39
|
Ong YS, Tang BL, Loo LS, Hong W. p125A exists as part of the mammalian Sec13/Sec31 COPII subcomplex to facilitate ER-Golgi transport. ACTA ACUST UNITED AC 2010; 190:331-45. [PMID: 20679433 PMCID: PMC2922642 DOI: 10.1083/jcb.201003005] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
p125A is an accessory protein for COPII-mediated vesicle budding that links the Sec13/Sec31 and Sec23/24 subcomplexes. Coat protein II (COPII)–mediated export from the endoplasmic reticulum (ER) involves sequential recruitment of COPII complex components, including the Sar1 GTPase, the Sec23/Sec24 subcomplex, and the Sec13/Sec31 subcomplex. p125A was originally identified as a Sec23A-interacting protein. Here we demonstrate that p125A also interacts with the C-terminal region of Sec31A. The Sec31A-interacting domain of p125A is between residues 260–600, and is therefore a distinct domain from that required for interaction with Sec23A. Gel filtration and immunodepletion studies suggest that the majority of cytosolic p125A exists as a ternary complex with the Sec13/Sec31A subcomplex, suggesting that Sec 13, Sec31A, and p125A exist in the cytosol primarily as preassembled Sec13/Sec31A/p125A heterohexamers. Golgi morphology and protein export from the ER were affected in p125A-silenced cells. Our results suggest that p125A is part of the Sec13/Sec31A subcomplex and facilitates ER export in mammalian cells.
Collapse
Affiliation(s)
- Yan Shan Ong
- Cancer and Developmental Cell Biology Division, Institute of Molecular and Cell Biology, Singapore 138673, Singapore
| | | | | | | |
Collapse
|
40
|
Regulation of Golgi structure and secretion by receptor-induced G protein βγ complex translocation. Proc Natl Acad Sci U S A 2010; 107:11417-22. [PMID: 20534534 DOI: 10.1073/pnas.1003042107] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We show that receptor induced G protein betagamma subunit translocation from the plasma membrane to the Golgi allows a receptor to initiate fragmentation and regulate secretion. A lung epithelial cell line, A549, was shown to contain an endogenous translocating G protein gamma subunit and exhibit receptor-induced Golgi fragmentation. Receptor-induced Golgi fragmentation was inhibited by a shRNA specific to the endogenous translocating gamma subunit. A kinase defective protein kinase D and a phospholipase C beta inhibitor blocked receptor-induced Golgi fragmentation, suggesting a role for this process in secretion. Consistent with betagamma translocation dependence, fragmentation induced by receptor activation was inhibited by a dominant negative nontranslocating gamma3. Insulin secretion was shown to be induced by muscarinic receptor activation in a pancreatic beta cell line, NIT-1. Induction of insulin secretion was also inhibited by the dominant negative gamma3 subunit consistent with the Golgi fragmentation induced by betagamma complex translocation playing a role in secretion.
Collapse
|
41
|
Hassinen A, Rivinoja A, Kauppila A, Kellokumpu S. Golgi N-glycosyltransferases form both homo- and heterodimeric enzyme complexes in live cells. J Biol Chem 2010; 285:17771-7. [PMID: 20378551 DOI: 10.1074/jbc.m110.103184] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glycans (i.e. oligosaccharide chains attached to cellular proteins and lipids) are crucial for nearly all aspects of life, including the development of multicellular organisms. They come in multiple forms, and much of this diversity between molecules, cells, and tissues is generated by Golgi-resident glycosidases and glycosyltransferases. However, their exact mode of functioning in glycan processing is currently unclear. Here we investigate the supramolecular organization of the N-glycosylation pathway in live cells by utilizing the bimolecular fluorescence complementation approach. We show that all four N-glycosylation enzymes tested (beta-1,2-N-acetylglucosaminyltransferase I, beta-1,2-N-acetylglucosaminyltransferase II, 1,4-galactosyltransferase I, and alpha-2,6-sialyltransferase I) form Golgi-localized homodimers. Intriguingly, the same enzymes also formed two distinct and functionally relevant heterodimers between the medial Golgi enzymes beta-1,2-N-acetylglucosaminyltransferase I and beta-1,2-N-acetylglucosaminyltransferase II and the trans-Golgi enzymes 1,4-galactosyltransferase I and alpha-2,6-sialyltransferase I. Given their strict Golgi localization and sequential order of function, the two heterodimeric complexes are probably responsible for the processing and maturation of N-glycans in live cells.
Collapse
Affiliation(s)
- Antti Hassinen
- Department of Biochemistry, University of Oulu and the Glycoscience Graduate School Finland, PO Box 3000, Linnanmaa, FIN-90014 Oulu, Finland
| | | | | | | |
Collapse
|
42
|
Manolea F, Chun J, Chen DW, Clarke I, Summerfeldt N, Dacks JB, Melançon P. Arf3 is activated uniquely at the trans-Golgi network by brefeldin A-inhibited guanine nucleotide exchange factors. Mol Biol Cell 2010; 21:1836-49. [PMID: 20357002 PMCID: PMC2877642 DOI: 10.1091/mbc.e10-01-0016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Arf3 associates with the TGN in a manner that is both temperature-sensitive and uniquely dependent on BIGs. TGN localization and release at 20°C are readily separated and depend on pairs of residues absolutely conserved and unique to Arf3 present at opposite ends of the protein. These results suggest that Arf3 plays a unique function at the TGN. It is widely assumed that class I and II Arfs function interchangeably throughout the Golgi complex. However, we report here that in vivo, Arf3 displays several unexpected properties. Unlike other Golgi-localized Arfs, Arf3 associates selectively with membranes of the trans-Golgi network (TGN) in a manner that is both temperature-sensitive and uniquely dependent on guanine nucleotide exchange factors of the BIGs family. For example, BIGs knockdown redistributed Arf3 but not Arf1 from Golgi membranes. Furthermore, shifting temperature to 20°C, a temperature known to block cargo in the TGN, selectively redistributed Arf3 from Golgi membranes. Arf3 redistribution occurred slowly, suggesting it resulted from a change in membrane composition. Arf3 knockdown and overexpression experiments suggest that redistribution is not responsible for the 20°C block. To investigate in more detail the mechanism for Arf3 recruitment and temperature-dependent release, we characterized several mutant forms of Arf3. This analysis demonstrated that those properties are readily separated and depend on pairs of residues present at opposite ends of the protein. Furthermore, phylogenetic analysis established that all four critical residues were absolutely conserved and unique to Arf3. These results suggest that Arf3 plays a unique function at the TGN that likely involves recruitment by a specific receptor.
Collapse
Affiliation(s)
- Florin Manolea
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Tu L, Banfield DK. Localization of Golgi-resident glycosyltransferases. Cell Mol Life Sci 2010; 67:29-41. [PMID: 19727557 PMCID: PMC11115592 DOI: 10.1007/s00018-009-0126-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 07/30/2009] [Accepted: 08/04/2009] [Indexed: 10/20/2022]
Abstract
For many glycosyltransferases, the information that instructs Golgi localization is located within a relatively short sequence of amino acids in the N-termini of these proteins comprising: the cytoplasmic tail, the transmembrane spanning region, and the stem region (CTS). Also, one enzyme may be more reliant on a particular region in the CTS for its localization than another. The predominance of these integral membrane proteins in the Golgi has seen these enzymes become central players in the development of membrane trafficking models of transport within this organelle. It is now understood that the means by which the characteristic distributions of glycosyltransferases arise within the subcompartments of the Golgi is inextricably linked to the mechanisms that cells employ to direct the flow of proteins and lipids within this organelle.
Collapse
Affiliation(s)
- Linna Tu
- Department of Biology, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, People’s Republic of China
| | - David Karl Banfield
- Department of Biology, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, People’s Republic of China
| |
Collapse
|
44
|
Ungar D. Golgi linked protein glycosylation and associated diseases. Semin Cell Dev Biol 2009; 20:762-9. [PMID: 19508859 DOI: 10.1016/j.semcdb.2009.03.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 03/10/2009] [Accepted: 03/11/2009] [Indexed: 11/17/2022]
Abstract
One of the Golgi's main functions is the glycosylation of secreted proteins. A large variety of glycan chains can be synthesized in the Golgi, and it is increasingly clear that these are critical in basic cellular functions as well as the development of multicellular organisms. The structurally best-documented glycans are N-glycans, yet these are also the most enigmatic in their function. In contrast, O-glycan function is far better understood, but here the structures and biosynthetic pathways are very incomplete. The critical importance of glycans is highlighted by the broad spectrum of diseases they are associated with, such as a number of inherited diseases, but also cancers or diabetes. The molecular clues to these, however, are only just being elucidated. Although some glycan structures are known to be involved in signaling or adhesion to the extracellular matrix, for most the functions are not yet known. This review aims at summarizing current knowledge as much as to point out critical areas key for future progress.
Collapse
Affiliation(s)
- Daniel Ungar
- University of York, Department of Biology (area 9), PO Box 373, York YO10 5YW, UK.
| |
Collapse
|
45
|
Rivinoja A, Hassinen A, Kokkonen N, Kauppila A, Kellokumpu S. Elevated Golgi pH impairs terminal N-glycosylation by inducing mislocalization of Golgi glycosyltransferases. J Cell Physiol 2009; 220:144-54. [PMID: 19277980 DOI: 10.1002/jcp.21744] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acidic pH of the Golgi lumen is known to be crucial for correct glycosylation, transport and sorting of proteins and lipids during their transit through the organelle. To better understand why Golgi acidity is important for these processes, we have examined here the most pH sensitive events in N-glycosylation by sequentially raising Golgi luminal pH with chloroquine (CQ), a weak base. We show that only a 0.2 pH unit increase (20 microM CQ) is sufficient to markedly impair terminal alpha(2,3)-sialylation of an N-glycosylated reporter protein (CEA), and to induce selective mislocalization of the corresponding alpha(2,3)-sialyltransferase (ST3) into the endosomal compartments. Much higher pH increase was required to impair alpha(2,6)-sialylation, or the proximal glycosylation steps such as beta(1,4)-galactosylation or acquisition of Endo H resistance, and the steady-state localization of the key enzymes responsible for these modifications (ST6, GalT I, MANII). The overall Golgi morphology also remained unaltered, except when Golgi pH was raised close to neutral. By using transmembrane domain chimeras between the ST6 and ST3, we also show that the luminal domain of the ST6 is mainly responsible for its less pH sensitive localization in the Golgi. Collectively, these results emphasize that moderate Golgi pH alterations such as those detected in cancer cells can impair N-glycosylation by inducing selective mislocalization of only certain Golgi glycosyltransferases.
Collapse
Affiliation(s)
- Antti Rivinoja
- Department of Biochemistry and The Finnish Glycoscience Graduate School, University of Oulu, Oulu, Finland
| | | | | | | | | |
Collapse
|
46
|
An HPLC-MALDI MS method for N-glycan analyses using smaller size samples: Application to monitor glycan modulation by medium conditions. Glycoconj J 2009; 26:1135-49. [DOI: 10.1007/s10719-009-9235-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 01/12/2009] [Accepted: 03/11/2009] [Indexed: 10/20/2022]
|
47
|
Codlin S, Mole SE. S. pombe btn1, the orthologue of the Batten disease gene CLN3, is required for vacuole protein sorting of Cpy1p and Golgi exit of Vps10p. J Cell Sci 2009; 122:1163-73. [PMID: 19299465 DOI: 10.1242/jcs.038323] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Batten disease is characterised by lysosomal dysfunction. The most common type of the disease is caused by mutations in the membrane protein CLN3, whose function is unknown. We show that the fission yeast orthologue Btn1p, previously implicated in vacuole function, is required for correct sorting of the vacuole hydrolase carboxypeptidase Y (Cpy1p). This is, in part, due to a defect in trafficking of Vps10p, the sorting receptor for Cpy1p, from the Golgi to the trans-Golgi network in btn1Delta cells. Our data also implicate btn1 in other Vps10-independent Cpy1-sorting pathways. Furthermore, btn1 affects the number, intracellular location and structure of Golgi compartments. We show that the prevacuole location of Btn1p is at the Golgi, because Btn1p colocalises predominantly with the Golgi marker Gms1p in compartments that are sensitive to Brefeldin A. Btn1p function might be linked to that of Vps34p, a phosphatidylinositol 3-kinase, because Btn1p acts as a multicopy suppressor of the severe Cpy1p vacuole protein-sorting defect of vps34Delta cells. Together, these results indicate an important role for Btn1p in the Golgi complex, which affects Golgi homeostasis and vacuole protein sorting. We propose a similar role for CLN3 in mammalian cells.
Collapse
Affiliation(s)
- Sandra Codlin
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
48
|
Kizuka Y, Tonoyama Y, Oka S. Distinct transport and intracellular activities of two GlcAT-P isoforms. J Biol Chem 2009; 284:9247-56. [PMID: 19181664 DOI: 10.1074/jbc.m807517200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A neural glycotope, human natural killer-1 carbohydrate, is involved in synaptic plasticity. The key biosynthetic enzyme is a glucuronyltransferase, GlcAT-P, a type II membrane protein comprising an N-terminal cytoplasmic tail, transmembrane domain, stem region, and C-terminal catalytic domain. Previously, we reported that GlcAT-P has two isoforms differing in only the presence or absence of the N-terminal 13 amino acids (P-N13) in the cytoplasmic tail, but the functional distinction of these two isoforms has not been reported. Herein, we show that when expressed in Neuro2A cells, short form GlcAT-P (sGlcAT-P) exhibited significantly higher glucuronylation activity than the longer form (lGlcAT-P), despite their comparable specific activities in vitro. In addition, sGlcAT-P was strictly localized in Golgi apparatus, whereas lGlcAT-P was mainly localized in Golgi but partly in the endoplasmic reticulum. We demonstrated that the small GTPase, Sar1, recognized a dibasic motif in the cytoplasmic tail near P-N13 that was important for exiting the endoplasmic reticulum, and Sar1 interacted with sGlcAT-P more strongly than lGlcAT-P. Finally, the attachment of P-N13 to another glycosyltransferase, polysialyltransferase-I (ST8Sia-IV), had similar effects, such as reduced activity and entrapment within endoplasmic reticulum. These results suggest that P-N13 can control glycosyltransferase transport through Sar1 binding interference.
Collapse
Affiliation(s)
- Yasuhiko Kizuka
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
49
|
Boucher R, Larkin H, Brodeur J, Gagnon H, Thériault C, Lavoie C. Intracellular trafficking of LRP9 is dependent on two acidic cluster/dileucine motifs. Histochem Cell Biol 2008; 130:315-27. [PMID: 18461348 DOI: 10.1007/s00418-008-0436-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2008] [Indexed: 01/09/2023]
Abstract
LDL receptor-related protein 9 (LRP9) is a distant member of the low-density lipoprotein receptor (LDLR) superfamily. To date, there are no reports on the cellular distribution of LRP9 or the signals responsible for its localization. Here, we investigated the intracellular localization and trafficking of LRP9. Using confocal microscopy, we demonstrated that LRP9 was not present at the plasma membrane but co-localized with various markers of the trans-Golgi network (TGN) and endosomes. This co-localization was dependent on the presence of two acidic cluster/dileucine (DXXLL) motifs in the cytoplasmic tail of LRP9, which interact with GGA proteins, clathrin adaptors involved in transport between the TGN and endosomes. LRP9 is the first example of a transmembrane protein with an internal GGA-binding sequence in addition to the usual C-terminal motif. An inactivating mutation (LL --> AA) in both DXXLL motifs, which completely inhibited the interaction of LRP9 with GGA proteins, led to an intracellular redistribution of LRP9 from the TGN to early endosomes and the cell surface, indicating that the two DXXLL motifs are essential sorting determinants of LRP9. In conclusion, our results suggest that LRP9 cycles between the TGN, endosomes and the plasma membrane through a GGA dependent-trafficking mechanism.
Collapse
Affiliation(s)
- Rémi Boucher
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001-12e Avenue Nord, Sherbrooke, QC, Canada, J1H 5N4
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Gangliosides are a family of glycolipids characterized by containing a variable number of sialic acid residues. Nearly, all animal cells contain at least some class of ganglioside in their membranes, but membranes from the CNS are characterized by their high content of these lipids. The synthesis of the oligosaccharide moiety of glycolipids is carried out in the Golgi complex. In this study, I will discuss the cellular and molecular basis of the organization of the glycosylating machinery in the Golgi complex, with particular attention to the mutual relationships, sub-Golgi localization, and intracellular trafficking of glycolipid glycosyltransferases, and to their relationships with the corresponding glycolipid acceptors and sugar nucleotide donors. I will also discuss how the organization of the glycosylating machinery in the Golgi may adapt to events controlling glycolipid expression.
Collapse
Affiliation(s)
- Hugo J F Maccioni
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC (UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, Argentina.
| |
Collapse
|