1
|
Pedroza LA, van den Haak F, Frumovitz A, Hernandez E, Hegewisch-Solloa E, Orange TK, Sheehan KB, Prockop S, Bodansky A, Chinn IK, Lupski JR, Posey JE, Mace EM, Li Y, Orange JS. The Golgi complex governs natural killer cell lytic granule positioning to promote directionality in cytotoxicity. Cell Rep 2025; 44:115156. [PMID: 39813120 PMCID: PMC11844255 DOI: 10.1016/j.celrep.2024.115156] [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: 03/14/2024] [Revised: 08/26/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cytotoxic immune cells mediate precise attacks against diseased cells to maintain organismal health. Their operational unit of killing and host defense is lytic granules (LGs), which are specialized lysosomal-related organelles. Precision in cytotoxicity is achieved by converging the many LGs to the microtubule-organizing center (MTOC) and polarizing these to the diseased cell for secretion. We identify unappreciated intimate relationships between the Golgi, MTOC, and LGs after cytotoxic cell activation, as well as the trans-Golgin protein GCC2 on the LG surface. GCC2 serves to tether LGs to the Golgi following convergence, and both GCC2 and the Golgi are required for the persistence of convergence. GCC2 allows LGs to utilize the Golgi as a docking station preventing LG dispersion and innocent bystander killing in complex three-dimensional environments. We also identify GCC2 variants causing human natural killer cell deficiency, further emphasizing the importance of LG convergence and Golgi linkage in precision targeting for human immunity.
Collapse
Affiliation(s)
- Luis A Pedroza
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Alexander Frumovitz
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Evelyn Hernandez
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Tabitha K Orange
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | - Aaron Bodansky
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ivan K Chinn
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Lupski
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emily M Mace
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yu Li
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jordan S Orange
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
2
|
Kim MS, Jeong H, Choi BH, Park J, Shin GS, Jung JH, Shin H, Kang KW, Jeon OH, Yu J, Park JH, Park Y, Choi Y, Kim HK, Hong S. GCC2 promotes non-small cell lung cancer progression by maintaining Golgi apparatus integrity and stimulating EGFR signaling pathways. Sci Rep 2024; 14:28926. [PMID: 39572606 PMCID: PMC11582359 DOI: 10.1038/s41598-024-75316-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/04/2024] [Indexed: 11/24/2024] Open
Abstract
Fundamental changes in intracellular processes, such as overactive growth signaling pathways, are common in carcinomas and are targets of many cancer therapeutics. GRIP and coiled-coil containing 2 (GCC2) is a trans-Golgi network (TGN) golgin maintaining Golgi apparatus structure and regulating vesicle transport. Here, we found an aberrant overexpression of GCC2 in non-small cell lung cancer (NSCLC) and conducted shRNA-mediated gene knockdown to investigate the role of GCC2 in NSCLC progression. shRNA-mediated GCC2 knockdown suppressed NSCLC cell growth, migration, stemness, and epithelial-mesenchymal transition (EMT) in vitro and tumor growth in vivo. In addition, GCC2 knockdown suppressed cancer cell exosome secretion and the oncogenic capacity of cancer cell-derived exosomes. Mechanistically, GCC2 inhibition decreased epidermal growth factor receptor (EGFR) expression and downstream growth and proliferation signaling. Furthermore, GCC2 inhibition compromised Golgi structural integrity in cancer cells, indicating a functional role of GCC2 in regulating intracellular trafficking and signaling to promote lung cancer progression. Together, these findings suggest GCC2 as a potential therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Min Sang Kim
- Department of Integrated Biomedical and Life Science, Graduate School of Korea University, Seoul, 02855, Republic of Korea
- BK21 FOUR R&E Center for Precision Public Health, Graduate School of Korea University, Seoul, 02855, Republic of Korea
| | - Hyesun Jeong
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Image Guided Precision Cancer Surgery Institute, Korea University, Seoul, 02841, Korea
| | - Jiho Park
- Department of Integrated Biomedical and Life Science, Graduate School of Korea University, Seoul, 02855, Republic of Korea
- BK21 FOUR R&E Center for Precision Public Health, Graduate School of Korea University, Seoul, 02855, Republic of Korea
| | - Gun Seop Shin
- Department of Integrated Biomedical and Life Science, Graduate School of Korea University, Seoul, 02855, Republic of Korea
- BK21 FOUR R&E Center for Precision Public Health, Graduate School of Korea University, Seoul, 02855, Republic of Korea
| | - Jik-Han Jung
- Department of Bio and Brain Engineeringand, KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hyunku Shin
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Ka-Won Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Ok Hwa Jeon
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Image Guided Precision Cancer Surgery Institute, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jewon Yu
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineeringand, KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Yong Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yeonho Choi
- BK21 FOUR R&E Center for Precision Public Health, Graduate School of Korea University, Seoul, 02855, Republic of Korea
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
- School of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Image Guided Precision Cancer Surgery Institute, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Sunghoi Hong
- Department of Integrated Biomedical and Life Science, Graduate School of Korea University, Seoul, 02855, Republic of Korea.
- BK21 FOUR R&E Center for Precision Public Health, Graduate School of Korea University, Seoul, 02855, Republic of Korea.
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
3
|
D’Souza Z, Sumya FT, Khakurel A, Lupashin V. Getting Sugar Coating Right! The Role of the Golgi Trafficking Machinery in Glycosylation. Cells 2021; 10:cells10123275. [PMID: 34943782 PMCID: PMC8699264 DOI: 10.3390/cells10123275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022] Open
Abstract
The Golgi is the central organelle of the secretory pathway and it houses the majority of the glycosylation machinery, which includes glycosylation enzymes and sugar transporters. Correct compartmentalization of the glycosylation machinery is achieved by retrograde vesicular trafficking as the secretory cargo moves forward by cisternal maturation. The vesicular trafficking machinery which includes vesicular coats, small GTPases, tethers and SNAREs, play a major role in coordinating the Golgi trafficking thereby achieving Golgi homeostasis. Glycosylation is a template-independent process, so its fidelity heavily relies on appropriate localization of the glycosylation machinery and Golgi homeostasis. Mutations in the glycosylation enzymes, sugar transporters, Golgi ion channels and several vesicle tethering factors cause congenital disorders of glycosylation (CDG) which encompass a group of multisystem disorders with varying severities. Here, we focus on the Golgi vesicle tethering and fusion machinery, namely, multisubunit tethering complexes and SNAREs and their role in Golgi trafficking and glycosylation. This review is a comprehensive summary of all the identified CDG causing mutations of the Golgi trafficking machinery in humans.
Collapse
|
4
|
Liang XH, Nichols JG, Hsu CW, Crooke ST. Hsc70 Facilitates Mannose-6-Phosphate Receptor-Mediated Intracellular Trafficking and Enhances Endosomal Release of Phosphorothioate-Modified Antisense Oligonucleotides. Nucleic Acid Ther 2021; 31:284-297. [PMID: 33567234 DOI: 10.1089/nat.2020.0920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phosphorothioate-modified antisense oligonucleotide (PS-ASO) drugs are commonly used to modulate gene expression through RNase H1-mediated cleavage of target RNAs. Upon internalization through endocytic pathways into cells, PS-ASOs must be released from membraned endosomal organelles to act on target RNAs, a limiting step of PS-ASO activity. Here we report that Hsc70 protein mediates productive release of PS-ASOs from endosomes. Hsc70 protein was enriched in endosome fractions shortly after PS-ASO incubation with cells. Reduction of Hsc70 significantly decreased the activities of PS-ASOs in reducing target RNAs. PS-ASO uptake and transport from early endosomes to late endosomes (LEs) were not affected upon Hsc70 reduction; however, endosomal release of PS-ASOs was impaired. Reduction of Hsc70 led to more scattered mannose-6-phosphate receptor (M6PR) localization at LEs in the cytoplasm, in contrast to the perinuclear localization at trans-Golgi network (TGN) in control cells, suggesting that retrograde transport of M6PR from LEs to TGN was affected. Consistently, reduction of Hsc70 increased colocalization of M6PR and PS-ASOs at LEs, and also delayed M6PR antibody transport from LE to TGN. Together, these results suggest that Hsc70 protein is involved in M6PR vesicle escape from LEs and may thus enhance PS-ASO release from LEs.
Collapse
Affiliation(s)
- Xue-Hai Liang
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Joshua G Nichols
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Chih-Wei Hsu
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Stanley T Crooke
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| |
Collapse
|
5
|
Zulkefli KL, Mahmoud IS, Williamson NA, Gosavi PK, Houghton FJ, Gleeson PA. A role for Rab30 in retrograde trafficking and maintenance of endosome-TGN organization. Exp Cell Res 2021; 399:112442. [PMID: 33359467 DOI: 10.1016/j.yexcr.2020.112442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 10/22/2022]
Abstract
Rab30 is a poorly characterized small GTPase. Here we show that Rab30 is localised primarily to the TGN and recycling endosomes in a range of cell types, including primary neurons; minor levels of Rab30 were also detected throughout the Golgi stack and early endosomes. Silencing of Rab30 resulted in the dispersal of both early and recycling endosomes and TGN compartments in HeLa cells. By analyzing cargo trafficking in Rab30-silenced and Rab30-overexpressing HeLa cells, we demonstrate that Rab30 plays a role in retrograde trafficking of TGN38 from endosomes to the Golgi, but has no apparent role in the endocytic recycling of the transferrin receptor to the plasma membrane. Five interactive partners with Rab30 were identified by pull-down and MS analysis using GFP-tagged Rab30 mutant, Rab30(Q68L). Two of the interactive partners identified were Arf1 and Arf4, known regulators of endosome to TGN retrograde transport. Knockdown of Arf1 and Arf4 results in GFP-Rab30 decorated tubules arising from the recycling endosomes, suggesting association of Rab30 with tubular carriers. Overall our data demonstrates a role for Rab30 in regulating retrograde transport to the TGN and maintenance of endosomal-TGN organization.
Collapse
Affiliation(s)
- Khalisah L Zulkefli
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Ismail S Mahmoud
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Nicholas A Williamson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia; The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Prajakta Kulkarni Gosavi
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Fiona J Houghton
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
6
|
Linders PTA, Peters E, ter Beest M, Lefeber DJ, van den Bogaart G. Sugary Logistics Gone Wrong: Membrane Trafficking and Congenital Disorders of Glycosylation. Int J Mol Sci 2020; 21:E4654. [PMID: 32629928 PMCID: PMC7369703 DOI: 10.3390/ijms21134654] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
Glycosylation is an important post-translational modification for both intracellular and secreted proteins. For glycosylation to occur, cargo must be transported after synthesis through the different compartments of the Golgi apparatus where distinct monosaccharides are sequentially bound and trimmed, resulting in increasingly complex branched glycan structures. Of utmost importance for this process is the intraorganellar environment of the Golgi. Each Golgi compartment has a distinct pH, which is maintained by the vacuolar H+-ATPase (V-ATPase). Moreover, tethering factors such as Golgins and the conserved oligomeric Golgi (COG) complex, in concert with coatomer (COPI) and soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-mediated membrane fusion, efficiently deliver glycosylation enzymes to the right Golgi compartment. Together, these factors maintain intra-Golgi trafficking of proteins involved in glycosylation and thereby enable proper glycosylation. However, pathogenic mutations in these factors can cause defective glycosylation and lead to diseases with a wide variety of symptoms such as liver dysfunction and skin and bone disorders. Collectively, this group of disorders is known as congenital disorders of glycosylation (CDG). Recent technological advances have enabled the robust identification of novel CDGs related to membrane trafficking components. In this review, we highlight differences and similarities between membrane trafficking-related CDGs.
Collapse
Affiliation(s)
- Peter T. A. Linders
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (P.T.A.L.); (E.P.); (M.t.B.)
| | - Ella Peters
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (P.T.A.L.); (E.P.); (M.t.B.)
| | - Martin ter Beest
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (P.T.A.L.); (E.P.); (M.t.B.)
| | - Dirk J. Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (P.T.A.L.); (E.P.); (M.t.B.)
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| |
Collapse
|
7
|
Liang XH, Sun H, Hsu CW, Nichols JG, Vickers TA, De Hoyos CL, Crooke ST. Golgi-endosome transport mediated by M6PR facilitates release of antisense oligonucleotides from endosomes. Nucleic Acids Res 2020; 48:1372-1391. [PMID: 31840180 PMCID: PMC7026651 DOI: 10.1093/nar/gkz1171] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022] Open
Abstract
Release of phosphorothioate antisense oligonucleotides (PS-ASOs) from late endosomes (LEs) is a rate-limiting step and a poorly defined process for productive intracellular ASO drug delivery. Here, we examined the role of Golgi-endosome transport, specifically M6PR shuttling mediated by GCC2, in PS-ASO trafficking and activity. We found that reduction in cellular levels of GCC2 or M6PR impaired PS-ASO release from endosomes and decreased PS-ASO activity in human cells. GCC2 relocated to LEs upon PS-ASO treatment, and M6PR also co-localized with PS-ASOs in LEs or on LE membranes. These proteins act through the same pathway to influence PS-ASO activity, with GCC2 action preceding that of M6PR. Our data indicate that M6PR binds PS-ASOs and facilitates their vesicular escape. The co-localization of M6PR and of GCC2 with ASOs is influenced by the PS modifications, which have been shown to enhance the affinity of ASOs for proteins, suggesting that localization of these proteins to LEs is mediated by ASO-protein interactions. Reduction of M6PR levels also decreased PS-ASO activity in mouse cells and in livers of mice treated subcutaneously with PS-ASO, indicating a conserved mechanism. Together, these results demonstrate that the transport machinery between LE and Golgi facilitates PS-ASO release.
Collapse
Affiliation(s)
- Xue-Hai Liang
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Hong Sun
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Chih-Wei Hsu
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Joshua G Nichols
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Timothy A Vickers
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Cheryl L De Hoyos
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| | - Stanley T Crooke
- Core Antisense Research, Ionis Pharmaceuticals, Inc. Carlsbad, CA 92104, USA
| |
Collapse
|
8
|
Fakhree MAA, Blum C, Claessens MMAE. Shaping membranes with disordered proteins. Arch Biochem Biophys 2019; 677:108163. [PMID: 31672499 DOI: 10.1016/j.abb.2019.108163] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/23/2019] [Accepted: 10/27/2019] [Indexed: 12/15/2022]
Abstract
Membrane proteins control and shape membrane trafficking processes. The role of protein structure in shaping cellular membranes is well established. However, a significant fraction of membrane proteins is disordered or contains long disordered regions. It becomes more and more clear that these disordered regions contribute to the function of membrane proteins. While the fold of a structured protein is essential for its function, being disordered seems to be a crucial feature of membrane bound intrinsically disordered proteins and protein regions. Here we outline the motifs that encode function in disordered proteins and discuss how these functional motifs enable disordered proteins to modulate membrane properties. These changes in membrane properties facilitate and regulate membrane trafficking processes which are highly abundant in eukaryotes.
Collapse
Affiliation(s)
| | - Christian Blum
- Nanobiophysics Group, University of Twente, 7522, NB, Enschede, the Netherlands
| | | |
Collapse
|
9
|
Cruz MD, Kim K. The inner workings of intracellular heterotypic and homotypic membrane fusion mechanisms. J Biosci 2019; 44:91. [PMID: 31502569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Intracellular trafficking is a field that has been intensively studied for years and yet there remains much to be learned. Part of the reason that there is so much obscurity remaining in this field is due to all the pathways and the stages that define cellular trafficking. One of the major steps in cellular trafficking is fusion. Fusion is defined as the terminal step that occurs when a cargo-laden vesicle arrives at the proper destination. There are two types of fusion within a cell: homotypic and heterotypic fusion. Homotypic fusion occurs when the two membranes merging together are of the same type such as vacuole to vacuole fusion. Heterotypic fusion occurs when the two membranes at play are of different types such as when an endosomal membrane fuses with a Golgi membrane. In this review, we will focus on all the protein components - Rabs, Golgins, Multisubunit tethers, GTPases, protein phosphatases and SNAREs - that have been known to function in both of these types of fusion. We hope to develop a model of how all of these constituents function together to achieve membrane fusion. Membrane fusion is a biological process absolutely necessary for proper intracellular trafficking. Due to the degree of importance multiple proteins are required for it to be properly carried through. Whether we are talking about heterotypic or homotypic fusion, any defects in the fusion machinery can result in disease states such as Parkinson's and Alzheimer's disease. Although much research has significantly expanded our knowledge of fusion, there is still much more to be learned.
Collapse
|
10
|
Delgado Cruz M, Kim K. The inner workings of intracellular heterotypic and homotypic membrane fusion mechanisms. J Biosci 2019. [DOI: 10.1007/s12038-019-9913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
11
|
Gillingham AK, Bertram J, Begum F, Munro S. In vivo identification of GTPase interactors by mitochondrial relocalization and proximity biotinylation. eLife 2019; 8:45916. [PMID: 31294692 PMCID: PMC6639074 DOI: 10.7554/elife.45916] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
The GTPases of the Ras superfamily regulate cell growth, membrane traffic and the cytoskeleton, and a wide range of diseases are caused by mutations in particular members. They function as switchable landmarks with the active GTP-bound form recruiting to the membrane a specific set of effector proteins. The GTPases are precisely controlled by regulators that promote acquisition of GTP (GEFs) or its hydrolysis to GDP (GAPs). We report here MitoID, a method for identifying effectors and regulators by performing in vivo proximity biotinylation with mitochondrially-localized forms of the GTPases. Applying this to 11 human Rab GTPases identified many known effectors and GAPs, as well as putative novel effectors, with examples of the latter validated for Rab2, Rab5, Rab9 and Rab11. MitoID can also efficiently identify effectors and GAPs of Rho and Ras family GTPases such as Cdc42, RhoA, Rheb, and N-Ras, and can identify GEFs by use of GDP-bound forms.
Collapse
Affiliation(s)
| | - Jessie Bertram
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Farida Begum
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
12
|
Ungermann C, Kümmel D. Structure of membrane tethers and their role in fusion. Traffic 2019; 20:479-490. [DOI: 10.1111/tra.12655] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/26/2019] [Accepted: 05/03/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Christian Ungermann
- Biochemistry Section, Department of Biology/ChemistryUniversity of Osnabrück Osnabrück Germany
- Center for Cellular Nanoanalytics (CellNanOs)University of Osnabrück Osnabrück Germany
| | - Daniel Kümmel
- Biochemistry & Structural Biology Section, Institute of BiochemistryUniversity of Münster Münster Germany
| |
Collapse
|
13
|
Abstract
The Golgi apparatus is a central intracellular membrane-bound organelle with key functions in trafficking, processing, and sorting of newly synthesized membrane and secretory proteins and lipids. To best perform these functions, Golgi membranes form a unique stacked structure. The Golgi structure is dynamic but tightly regulated; it undergoes rapid disassembly and reassembly during the cell cycle of mammalian cells and is disrupted under certain stress and pathological conditions. In the past decade, significant amount of effort has been made to reveal the molecular mechanisms that regulate the Golgi membrane architecture and function. Here we review the major discoveries in the mechanisms of Golgi structure formation, regulation, and alteration in relation to its functions in physiological and pathological conditions to further our understanding of Golgi structure and function in health and diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Abstract
The role of the Golgi apparatus in carcinogenesis still remains unclear. A number of structural and functional cis-, medial-, and trans-Golgi proteins as well as a complexity of metabolic pathways which they mediate may indicate a central role of the Golgi apparatus in the development and progression of cancer. Pleiotropy of cellular function of the Golgi apparatus makes it a "metabolic heart" or a relay station of a cell, which combines multiple signaling pathways involved in carcinogenesis. Therefore, any damage to or structural abnormality of the Golgi apparatus, causing its fragmentation and/or biochemical dysregulation, results in an up- or downregulation of signaling pathways and may in turn promote tumor progression, as well as local nodal and distant metastases. Three alternative or parallel models of spatial and functional Golgi organization within tumor cells were proposed: (1) compacted Golgi structure, (2) normal Golgi structure with its increased activity, and (3) the Golgi fragmentation with ministacks formation. Regardless of the assumed model, the increased activity of oncogenesis initiators and promoters with inhibition of suppressor proteins results in an increased cell motility and migration, increased angiogenesis, significantly activated trafficking kinetics, proliferation, EMT induction, decreased susceptibility to apoptosis-inducing factors, and modulating immune response to tumor cell antigens. Eventually, this will lead to the increased metastatic potential of cancer cells and an increased risk of lymph node and distant metastases. This chapter provided an overview of the current state of knowledge of selected Golgi proteins, their role in cytophysiology as well as potential involvement in tumorigenesis.
Collapse
|
15
|
Renna L, Stefano G, Slabaugh E, Wormsbaecher C, Sulpizio A, Zienkiewicz K, Brandizzi F. TGNap1 is required for microtubule-dependent homeostasis of a subpopulation of the plant trans-Golgi network. Nat Commun 2018. [PMID: 30552321 DOI: 10.1038/s41467-018-07662-7664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
Defining convergent and divergent mechanisms underlying the biogenesis and function of endomembrane organelles is fundamentally important in cell biology. In all eukaryotes, the Trans-Golgi Network (TGN) is the hub where the exocytic and endocytic pathways converge. To gain knowledge in the mechanisms underlying TGN biogenesis and function, we characterized TGNap1, a protein encoded by a plant gene of unknown function conserved with metazoans. We demonstrate that TGNap1 is a TGN protein required for the homeostasis of biosynthetic and endocytic traffic pathways. We also show that TGNap1 binds Rab6, YIP4 and microtubules. Finally, we establish that TGNap1 contributes to microtubule-dependent biogenesis, tracking and function of a TGN subset, likely through interaction with Rab6 and YIP4. Our results identify an important trafficking determinant at the plant TGN and reveal an unexpected reliance of post-Golgi traffic homeostasis and organelle biogenesis on microtubules in plants.
Collapse
Affiliation(s)
- Luciana Renna
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
| | - Giovanni Stefano
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Plant Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Erin Slabaugh
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Clarissa Wormsbaecher
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Alan Sulpizio
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14850, USA
| | - Krzysztof Zienkiewicz
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Plant Biochemistry, Georg-August-University, Albrecht-von-Haller-Institute for Plant Sciences, 37073, Göttingen, Germany
| | - Federica Brandizzi
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Plant Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
16
|
Renna L, Stefano G, Slabaugh E, Wormsbaecher C, Sulpizio A, Zienkiewicz K, Brandizzi F. TGNap1 is required for microtubule-dependent homeostasis of a subpopulation of the plant trans-Golgi network. Nat Commun 2018; 9:5313. [PMID: 30552321 PMCID: PMC6294250 DOI: 10.1038/s41467-018-07662-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/08/2018] [Indexed: 11/09/2022] Open
Abstract
Defining convergent and divergent mechanisms underlying the biogenesis and function of endomembrane organelles is fundamentally important in cell biology. In all eukaryotes, the Trans-Golgi Network (TGN) is the hub where the exocytic and endocytic pathways converge. To gain knowledge in the mechanisms underlying TGN biogenesis and function, we characterized TGNap1, a protein encoded by a plant gene of unknown function conserved with metazoans. We demonstrate that TGNap1 is a TGN protein required for the homeostasis of biosynthetic and endocytic traffic pathways. We also show that TGNap1 binds Rab6, YIP4 and microtubules. Finally, we establish that TGNap1 contributes to microtubule-dependent biogenesis, tracking and function of a TGN subset, likely through interaction with Rab6 and YIP4. Our results identify an important trafficking determinant at the plant TGN and reveal an unexpected reliance of post-Golgi traffic homeostasis and organelle biogenesis on microtubules in plants.
Collapse
Affiliation(s)
- Luciana Renna
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
| | - Giovanni Stefano
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Plant Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Erin Slabaugh
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Clarissa Wormsbaecher
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Alan Sulpizio
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14850, USA
| | - Krzysztof Zienkiewicz
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA
- Department of Plant Biochemistry, Georg-August-University, Albrecht-von-Haller-Institute for Plant Sciences, 37073, Göttingen, Germany
| | - Federica Brandizzi
- MSU-DOE Plant Research Lab, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Plant Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
17
|
Makaraci P, Kim K. trans-Golgi network-bound cargo traffic. Eur J Cell Biol 2018; 97:137-149. [PMID: 29398202 DOI: 10.1016/j.ejcb.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Cargo following the retrograde trafficking are sorted at endosomes to be targeted the trans-Golgi network (TGN), a central receiving organelle. Though molecular requirements and their interaction networks have been somewhat established, the complete understanding of the intricate nature of their action mechanisms in every step of the retrograde traffic pathway remains unachieved. This review focuses on elucidating known functions of key regulators, including scission factors at the endosome and tethering/fusion mediators at the receiving dock, TGN, as well as a diverse range of cargo.
Collapse
Affiliation(s)
- Pelin Makaraci
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA.
| |
Collapse
|
18
|
Abstract
GTP-ases of the Rab family (about 70 in human) are key regulators of intracellular transport and membrane trafficking in eukaryotic cells. Remarkably, almost one third associate with membranes of the Golgi complex and TGN (trans-Golgi network). Through interactions with a variety of effectors that include molecular motors, tethering complexes, scaffolding proteins and lipid kinases, they play an important role in maintaining Golgi architecture.
Collapse
Affiliation(s)
- Bruno Goud
- a Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport , Paris , France
| | - Shijie Liu
- b Department of Physiology and Biophysics , University of Arkansas for Medical Sciences , Little Rock , USA
| | - Brian Storrie
- b Department of Physiology and Biophysics , University of Arkansas for Medical Sciences , Little Rock , USA
| |
Collapse
|
19
|
Tzeng HT, Li TH, Tang YA, Tsai CH, Frank Lu PJ, Lai WW, Chiang CW, Wang YC. Phosphorylation of Rab37 by protein kinase C alpha inhibits the exocytosis function and metastasis suppression activity of Rab37. Oncotarget 2017; 8:108556-108570. [PMID: 29312551 PMCID: PMC5752464 DOI: 10.18632/oncotarget.20998] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/05/2017] [Indexed: 01/31/2023] Open
Abstract
We previously identified a novel Rab small GTPase protein, Rab37, which plays a critical role in regulating exocytosis of secreted glycoproteins, tissue inhibitor of metalloproteinases 1 (TIMP1) to suppress lung cancer metastasis. Patients with preserved Rab37 protein expression were associated with better prognosis. However, a significant number of the patients with preserved Rab37 expression showed poor survival. In addition, the molecular mechanism for the regulation of Rab37-mediated exocytosis remained to be further identified. Therefore, we investigated the molecular mechanism underlying the dysregulation of Rab37-mediated exocytosis and metastasis suppression. Here, we report a novel mechanism for Rab37 inactivation by phosphorylation. Lung cancer patients with preserved Rab37, low TIMP1, and high PKCα expression profile correlate with worse progression-free survival examined by Kaplan-Meier survival, suggesting that PKCα overexpression leads to dysfunction of Rab37. This PKCα-Rab37-TIMP1 expression profile predicts the poor outcome by multivariate Cox regression analysis. We also show that Rab37 is phosphorylated by protein kinase Cα (PKCα) at threonine 172 (T172), leading to attenuation of its GTP-bound state, and impairment of the Rab37-mediated exocytosis of TIMP1, and thus reduces its suppression activity on lung cancer cell motility. We further demonstrate that PKCα reduces vesicle colocalization of Rab37 and TIMP1, and therefore inhibits Rab37-mediated TIMP1 trafficking. Moreover, Phospho-mimetic aspartate substitution mutant T172D of Rab37 significantly promotes tumor metastasis in vivo. Our findings reveal a novel regulation of Rab37 activity by PKCα-mediated phosphorylation which inhibits exocytic transport of TIMP1 and thereby enhances lung tumor metastasis.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Hsin Li
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-An Tang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology and Research (ASTAR), Singapore
| | - Chung-Han Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jung Frank Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Wei Lai
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Wu Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
20
|
Abstract
ADP-ribosylation factors (Arfs) and ADP-ribosylation factor-like proteins (Arls) are highly conserved small GTPases that function as main regulators of vesicular trafficking and cytoskeletal reorganization. Arl1, the first identified member of the large Arl family, is an important regulator of Golgi complex structure and function in organisms ranging from yeast to mammals. Together with its effectors, Arl1 has been shown to be involved in several cellular processes, including endosomal trans-Golgi network and secretory trafficking, lipid droplet and salivary granule formation, innate immunity and neuronal development, stress tolerance, as well as the response of the unfolded protein. In this Commentary, we provide a comprehensive summary of the Arl1-dependent cellular functions and a detailed characterization of several Arl1 effectors. We propose that involvement of Arl1 in these diverse cellular functions reflects the fact that Arl1 is activated at several late-Golgi sites, corresponding to specific molecular complexes that respond to and integrate multiple signals. We also provide insight into how the GTP-GDP cycle of Arl1 is regulated, and highlight a newly discovered mechanism that controls the sophisticated regulation of Arl1 activity at the Golgi complex.
Collapse
Affiliation(s)
- Chia-Jung Yu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Linkou, Tao-Yuan 33302, Taiwan.,Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Tao-Yuan 33305, Taiwan
| | - Fang-Jen S Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan .,Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| |
Collapse
|
21
|
Saimani U, Kim K. Traffic from the endosome towards trans-Golgi network. Eur J Cell Biol 2017; 96:198-205. [PMID: 28256269 DOI: 10.1016/j.ejcb.2017.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/24/2017] [Accepted: 02/16/2017] [Indexed: 11/16/2022] Open
Abstract
Retrograde passage of a transport carrier entails cargo sorting at the endosome, generation of a cargo-laden carrier and its movement along cytoskeletal tracks towards trans-Golgi network (TGN), tethering at the TGN, and fusion with the Golgi membrane. Significant advances have been made in understanding this traffic system, revealing molecular requirements in each step and the functional connection between them as well as biomedical implication of the dysregulation of those important traffic factors. This review focuses on describing up-to-date action mechanisms for retrograde transport from the endosomal system to the TGN.
Collapse
Affiliation(s)
- Uma Saimani
- Department of Biology, Missouri State University, 901 S National, Springfield, MO 65807, United States
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National, Springfield, MO 65807, United States.
| |
Collapse
|
22
|
Abstract
Rab proteins regulate vesicular transport in eukaryotic cells and establish connections to various cellular structures and processes by interacting with so-called effector molecules. Several of these effectors are known to not only bind a single Rab protein, but to be able to bind multiple different Rabs simultaneously. In this review we will give a short overview of effectors in general and (putative) functions of the aforementioned multivalent Rab:effector interactions.
Collapse
Affiliation(s)
- Amrita Rai
- a Department of Structural Biochemistry , Max Planck Institute of Molecular Physiology , Dortmund , Germany
| | - Roger S Goody
- a Department of Structural Biochemistry , Max Planck Institute of Molecular Physiology , Dortmund , Germany
| | - Matthias P Müller
- a Department of Structural Biochemistry , Max Planck Institute of Molecular Physiology , Dortmund , Germany
| |
Collapse
|
23
|
Wong M, Gillingham AK, Munro S. The golgin coiled-coil proteins capture different types of transport carriers via distinct N-terminal motifs. BMC Biol 2017; 15:3. [PMID: 28122620 PMCID: PMC5267433 DOI: 10.1186/s12915-016-0345-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022] Open
Abstract
Background The internal organization of cells depends on mechanisms to ensure that transport carriers, such as vesicles, fuse only with the correct destination organelle. Several types of proteins have been proposed to confer specificity to this process, and we have recently shown that a set of coiled-coil proteins on the Golgi, called golgins, are able to capture specific classes of carriers when relocated to an ectopic location. Results Mapping of six different golgins reveals that, in each case, a short 20–50 residue region is necessary and sufficient to capture specific carriers. In all six of GMAP-210, golgin-84, TMF, golgin-97, golgin-245, and GCC88, this region is located at the extreme N-terminus of the protein. The vesicle-capturing regions of GMAP-210, golgin-84, and TMF capture intra-Golgi vesicles and share some sequence features, suggesting that they act in a related, if distinct, manner. In the case of GMAP-210, this shared feature is in addition to a previously characterized “amphipathic lipid-packing sensor” motif that can capture highly curved membranes, with the two motifs being apparently involved in capturing distinct types of vesicles. Of the three GRIP domain golgins that capture endosome-to-Golgi carriers, golgin-97 and golgin-245 share a closely related capture motif, whereas that in GCC88 is distinct, suggesting that it works by a different mechanism and raising the possibility that the three golgins capture different classes of endosome-derived carriers that share many cargos but have distinct features for recognition at the Golgi. Conclusions For six different golgins, the capture of carriers is mediated by a short region at the N-terminus of the protein. There appear to be at least four different types of motif, consistent with specific golgins capturing specific classes of carrier and implying the existence of distinct receptors present on each of these different carrier classes.
Collapse
Affiliation(s)
- Mie Wong
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Alison K Gillingham
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
24
|
Rai A, Oprisko A, Campos J, Fu Y, Friese T, Itzen A, Goody RS, Gazdag EM, Müller MP. bMERB domains are bivalent Rab8 family effectors evolved by gene duplication. eLife 2016; 5. [PMID: 27552051 PMCID: PMC5026484 DOI: 10.7554/elife.18675] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022] Open
Abstract
In their active GTP-bound form, Rab proteins interact with proteins termed effector molecules. In this study, we have thoroughly characterized a Rab effector domain that is present in proteins of the Mical and EHBP families, both known to act in endosomal trafficking. Within our study, we show that these effectors display a preference for Rab8 family proteins (Rab8, 10, 13 and 15) and that some of the effector domains can bind two Rab proteins via separate binding sites. Structural analysis allowed us to explain the specificity towards Rab8 family members and the presence of two similar Rab binding sites that must have evolved via gene duplication. This study is the first to thoroughly characterize a Rab effector protein that contains two separate Rab binding sites within a single domain, allowing Micals and EHBPs to bind two Rabs simultaneously, thus suggesting previously unknown functions of these effector molecules in endosomal trafficking. DOI:http://dx.doi.org/10.7554/eLife.18675.001
Collapse
Affiliation(s)
- Amrita Rai
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Anastasia Oprisko
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Jeremy Campos
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Yangxue Fu
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Timon Friese
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Aymelt Itzen
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Garching, Germany
| | - Roger S Goody
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Emerich Mihai Gazdag
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Matthias P Müller
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| |
Collapse
|
25
|
Abstract
Intracellular membrane fusion is mediated in most cases by membrane-bridging complexes of soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). However, the assembly of such complexes in vitro is inefficient, and their uncatalysed disassembly is undetectably slow. Here, we focus on the cellular machinery that orchestrates assembly and disassembly of SNARE complexes, thereby regulating processes ranging from vesicle trafficking to organelle fusion to neurotransmitter release. Rapid progress is being made on many fronts, including the development of more realistic cell-free reconstitutions, the application of single-molecule biophysics, and the elucidation of X-ray and high-resolution electron microscopy structures of the SNARE assembly and disassembly machineries 'in action'.
Collapse
Affiliation(s)
- Richard W Baker
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA.,Present address: Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Frederick M Hughson
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
26
|
Kumar M, Kaur S, Nazir A, Tripathi RK. HIV-1 Nef binds with human GCC185 protein and regulates mannose 6 phosphate receptor recycling. Biochem Biophys Res Commun 2016; 474:137-145. [DOI: 10.1016/j.bbrc.2016.04.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/18/2016] [Indexed: 01/28/2023]
|
27
|
Cheung PYP, Pfeffer SR. Transport Vesicle Tethering at the Trans Golgi Network: Coiled Coil Proteins in Action. Front Cell Dev Biol 2016; 4:18. [PMID: 27014693 PMCID: PMC4791371 DOI: 10.3389/fcell.2016.00018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022] Open
Abstract
The Golgi complex is decorated with so-called Golgin proteins that share a common feature: a large proportion of their amino acid sequences are predicted to form coiled-coil structures. The possible presence of extensive coiled coils implies that these proteins are highly elongated molecules that can extend a significant distance from the Golgi surface. This property would help them to capture or trap inbound transport vesicles and to tether Golgi mini-stacks together. This review will summarize our current understanding of coiled coil tethers that are needed for the receipt of transport vesicles at the trans Golgi network (TGN). How do long tethering proteins actually catch vesicles? Golgi-associated, coiled coil tethers contain numerous binding sites for small GTPases, SNARE proteins, and vesicle coat proteins. How are these interactions coordinated and are any or all of them important for the tethering process? Progress toward understanding these questions and remaining, unresolved mysteries will be discussed.
Collapse
Affiliation(s)
- Pak-Yan P Cheung
- Department of Biochemistry, Stanford University School of Medicine Stanford, CA, USA
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
28
|
Finding the Golgi: Golgin Coiled-Coil Proteins Show the Way. Trends Cell Biol 2016; 26:399-408. [PMID: 26972448 DOI: 10.1016/j.tcb.2016.02.005] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 01/27/2023]
Abstract
The Golgi apparatus lies at the centre of the secretory pathway. It consists of a series of flattened compartments typically organised into a stack that, in mammals, is connected to additional stacks to form a Golgi ribbon. The Golgi is responsible for the maturation and modification of proteins and lipids, and receives and exports vesicles to and from multiple destinations within the cell. This complex trafficking network requires that only the correct vesicles fuse with the correct destination membrane. Recently, a group of coiled-coil proteins called golgins were shown to not only capture incoming vesicles but to also provide specificity to the tethering step. This raises many interesting questions about how they interact with other components of membrane traffic, some of which may also contribute to specificity.
Collapse
|
29
|
Witkos TM, Lowe M. The Golgin Family of Coiled-Coil Tethering Proteins. Front Cell Dev Biol 2016; 3:86. [PMID: 26793708 PMCID: PMC4707255 DOI: 10.3389/fcell.2015.00086] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/14/2015] [Indexed: 01/28/2023] Open
Abstract
The golgins are a family of predominantly coiled-coil proteins that are localized to the Golgi apparatus. Golgins are present in all eukaryotes, suggesting an evolutionary conserved function. Golgins are anchored to the Golgi membrane by their carboxy terminus and are predicted to adopt an extended conformation that projects into the surrounding cytoplasm. This arrangement is ideal for the capture or tethering of nearby membranes or cytoskeletal elements. Golgin-mediated tethering is thought to be important for vesicular traffic at the Golgi apparatus, the maintenance of Golgi architecture, as well as the positioning of the Golgi apparatus within cells. In addition to acting as tethers, some golgins can also sequester various factors at the Golgi membrane, allowing for the spatiotemporal regulation of downstream cellular functions. Although it is now established that golgins are membrane and cytoskeleton tethers, the mechanisms underlying tethering remain poorly defined. Moreover, the importance of golgin-mediated tethering in a physiological context remains to be fully explored. This review will describe our current understanding of golgin function, highlighting recent progress that has been made, and goes on to discuss outstanding questions and potential avenues for future research with regard to this family of conserved Golgi-associated proteins.
Collapse
Affiliation(s)
- Tomasz M Witkos
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | - Martin Lowe
- Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
30
|
Cheung PYP, Limouse C, Mabuchi H, Pfeffer SR. Protein flexibility is required for vesicle tethering at the Golgi. eLife 2015; 4. [PMID: 26653856 PMCID: PMC4721967 DOI: 10.7554/elife.12790] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/13/2015] [Indexed: 01/27/2023] Open
Abstract
The Golgi is decorated with coiled-coil proteins that may extend long distances to help vesicles find their targets. GCC185 is a trans Golgi-associated protein that captures vesicles inbound from late endosomes. Although predicted to be relatively rigid and highly extended, we show that flexibility in a central region is required for GCC185’s ability to function in a vesicle tethering cycle. Proximity ligation experiments show that that GCC185’s N-and C-termini are within <40 nm of each other on the Golgi. In physiological buffers without fixatives, atomic force microscopy reveals that GCC185 is shorter than predicted, and its flexibility is due to a central bubble that represents local unwinding of specific sequences. Moreover, 85% of the N-termini are splayed, and the splayed N-terminus can capture transport vesicles in vitro. These unexpected features support a model in which GCC185 collapses onto the Golgi surface, perhaps by binding to Rab GTPases, to mediate vesicle tethering.
Collapse
Affiliation(s)
| | - Charles Limouse
- Department of Applied Physics, Stanford University, Stanford, United States
| | - Hideo Mabuchi
- Department of Applied Physics, Stanford University, Stanford, United States
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
31
|
Cheung PYP, Pfeffer SR. Molecular and cellular characterization of GCC185: a tethering protein of the trans-Golgi network. Methods Mol Biol 2015; 1270:179-90. [PMID: 25702118 DOI: 10.1007/978-1-4939-2309-0_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Transport vesicle tethers are proteins that link partner membranes together to permit subsequent SNARE protein pairing and fusion. Despite the identification of a relatively large number of tethering proteins, little is known about the precise mechanisms by which they act. Biochemical isolation of tethers permits direct analysis of their physical characteristics and molecular interactions. Here, we describe the expression and purification of GCC185, a trans-Golgi-localized, 190-kDa coiled-coil tethering protein. In addition, we present a gene rescue approach to analyze the function of this tether after its depletion from cells using siRNA.
Collapse
Affiliation(s)
- Pak-Yan Patricia Cheung
- Department of Biochemistry, Stanford University School of Medicine, Beckman Center, Stanford University, Stanford, CA, 94305-5307, USA
| | | |
Collapse
|
32
|
Aizawa M, Fukuda M. Small GTPase Rab2B and Its Specific Binding Protein Golgi-associated Rab2B Interactor-like 4 (GARI-L4) Regulate Golgi Morphology. J Biol Chem 2015. [PMID: 26209634 DOI: 10.1074/jbc.m115.669242] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rab small GTPases are crucial regulators of the membrane traffic that maintains organelle identity and morphology. Several Rab isoforms are present in the Golgi, and it has been suggested that they regulate the compacted morphology of the Golgi in mammalian cells. However, the functional relationships among the Golgi-resident Rabs, e.g. whether they are functionally redundant or different, are poorly understood. In this study, we used specific siRNAs to perform genome-wide screening for human Rabs that are involved in Golgi morphology in HeLa-S3 cells. The results showed that knockdown of any one of the six Rab isoforms (Rab1A/1B/2A/2B/6B/8A) induced fragmentation of the Golgi in HeLa-S3 cells and that its phenotype was rescued by re-expression of their respective siRNA-resistant construct. We then performed systematic knockdown-rescue experiments in relation to each of the six Rabs. Interestingly, with the exception of the Rab8A knockdown, the Golgi fragmentation phenotype induced by knockdown of a single Rab isoform, e.g. Rab2B, was efficiently rescued by re-expression of its siRNA-resistant Rab alone, not by any of the other five Rabs, e.g. Rab2A, which is highly homologous to Rab2B, indicating that these Rab isoforms non-redundantly regulate Golgi morphology possibly through interaction with isoform-specific effector molecules. In addition, we identified Golgi-associated Rab2B interactor-like 4 (GARI-L4) as a novel Golgi-resident Rab2B-specific binding protein whose knockdown also induced fragmentation of the Golgi. Our findings suggest that the compacted Golgi morphology of mammalian cells is finely tuned by multiple sets of Rab (or Rab-effector complexes) that for the most part function independently.
Collapse
Affiliation(s)
- Megumi Aizawa
- From the Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- From the Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
33
|
Egerer J, Emmerich D, Fischer-Zirnsak B, Chan WL, Meierhofer D, Tuysuz B, Marschner K, Sauer S, Barr FA, Mundlos S, Kornak U. GORAB Missense Mutations Disrupt RAB6 and ARF5 Binding and Golgi Targeting. J Invest Dermatol 2015; 135:2368-2376. [PMID: 26000619 DOI: 10.1038/jid.2015.192] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 11/09/2022]
Abstract
Gerodermia osteodysplastica is a hereditary segmental progeroid disorder affecting skin, connective tissues, and bone that is caused by loss-of-function mutations in GORAB. The golgin, RAB6-interacting (GORAB) protein localizes to the Golgi apparatus and interacts with the small GTPase RAB6. In this study, we used different approaches to shed more light on the recruitment of GORAB to this compartment. We show that GORAB best colocalizes with trans-Golgi markers and is rapidly displaced upon Brefeldin A exposition, indicating a loose association with Golgi membranes. A yeast two-hybrid screening revealed a specific interaction with the small GTPase ADP-ribosylation factor (ARF5) in its active, GTP-bound form. ARF5 and RAB6 bind to GORAB via the same internal Golgi-targeting RAB6 and ARF5 binding (IGRAB) domain. Two GORAB missense mutations identified in gerodermia osteodysplastica patients fall within this IGRAB domain. GORAB carrying the mutation p.Ala220Pro had a cytoplasmic distribution and failed to interact with both RAB6 and ARF5. In contrast, the p.Ser175Phe mutation displaced GORAB from the Golgi compartment to vesicular structures and selectively impaired ARF5 binding. Our findings indicate that the IGRAB domain is crucial for the Golgi localization of GORAB and that loss of this localization impairs its physiological function.
Collapse
Affiliation(s)
- Johannes Egerer
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Denise Emmerich
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Björn Fischer-Zirnsak
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - Wing Lee Chan
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany
| | - David Meierhofer
- Max-Planck-Institut fuer Molekulare Genetik, Mass Spectrometry Facility, Berlin, Germany
| | - Beyhan Tuysuz
- Department of Pediatric Genetics, Cerrahpaşa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Katrin Marschner
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Sascha Sauer
- Max-Planck-Institut fuer Molekulare Genetik, Otto-Warburg-Laboratories, Berlin, Germany
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Stefan Mundlos
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Uwe Kornak
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany; Max-Planck-Institut fuer Molekulare Genetik, FG Development & Disease, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitaetsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
34
|
Sato K, Roboti P, Mironov AA, Lowe M. Coupling of vesicle tethering and Rab binding is required for in vivo functionality of the golgin GMAP-210. Mol Biol Cell 2014; 26:537-53. [PMID: 25473115 PMCID: PMC4310744 DOI: 10.1091/mbc.e14-10-1450] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vesicle tethering mediated by the golgin GMAP-210 is required to maintain the structure of the Golgi apparatus. Tethering by GMAP-210 is mediated solely by the ALPS motif, and binding to Rab2 and the length of GMAP-210, although not required for tethering per se, are also critical for its functional role at the Golgi apparatus. Golgins are extended coiled-coil proteins believed to participate in membrane-tethering events at the Golgi apparatus. However, the importance of golgin-mediated tethering remains poorly defined, and alternative functions for golgins have been proposed. Moreover, although golgins bind to Rab GTPases, the functional significance of Rab binding has yet to be determined. In this study, we show that depletion of the golgin GMAP-210 causes a loss of Golgi cisternae and accumulation of numerous vesicles. GMAP-210 function in vivo is dependent upon its ability to tether membranes, which is mediated exclusively by the amino-terminal ALPS motif. Binding to Rab2 is also important for GMAP-210 function, although it is dispensable for tethering per se. GMAP-210 length is also functionally important in vivo. Together our results indicate a key role for GMAP-210–mediated membrane tethering in maintaining Golgi structure and support a role for Rab2 binding in linking tethering with downstream docking and fusion events at the Golgi apparatus.
Collapse
Affiliation(s)
- Keisuke Sato
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Peristera Roboti
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Alexander A Mironov
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Martin Lowe
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
35
|
Wandinger-Ness A, Zerial M. Rab proteins and the compartmentalization of the endosomal system. Cold Spring Harb Perspect Biol 2014; 6:a022616. [PMID: 25341920 PMCID: PMC4413231 DOI: 10.1101/cshperspect.a022616;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Of the approximately 70 human Rab GTPases, nearly three-quarters are involved in endocytic trafficking. Significant plasticity in endosomal membrane transport pathways is closely coupled to receptor signaling and Rab GTPase-regulated scaffolds. Here we review current literature pertaining to endocytic Rab GTPase localizations, functions, and coordination with regulatory proteins and effectors. The roles of Rab GTPases in (1) compartmentalization of the endocytic pathway into early, recycling, late, and lysosomal routes; (2) coordination of individual transport steps from vesicle budding to fusion; (3) effector interactomes; and (4) integration of GTPase and signaling cascades are discussed.
Collapse
Affiliation(s)
- Angela Wandinger-Ness
- Department of Pathology MSC08 4640, University of New Mexico HSC, Albuquerque, New Mexico 87131
| | - Marino Zerial
- Max Planck Institute of Molecular and Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
36
|
Wandinger-Ness A, Zerial M. Rab proteins and the compartmentalization of the endosomal system. Cold Spring Harb Perspect Biol 2014; 6:a022616. [PMID: 25341920 DOI: 10.1101/cshperspect.a022616] [Citation(s) in RCA: 429] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Of the approximately 70 human Rab GTPases, nearly three-quarters are involved in endocytic trafficking. Significant plasticity in endosomal membrane transport pathways is closely coupled to receptor signaling and Rab GTPase-regulated scaffolds. Here we review current literature pertaining to endocytic Rab GTPase localizations, functions, and coordination with regulatory proteins and effectors. The roles of Rab GTPases in (1) compartmentalization of the endocytic pathway into early, recycling, late, and lysosomal routes; (2) coordination of individual transport steps from vesicle budding to fusion; (3) effector interactomes; and (4) integration of GTPase and signaling cascades are discussed.
Collapse
Affiliation(s)
- Angela Wandinger-Ness
- Department of Pathology MSC08 4640, University of New Mexico HSC, Albuquerque, New Mexico 87131
| | - Marino Zerial
- Max Planck Institute of Molecular and Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
37
|
Abstract
Membrane trafficking depends on transport vesicles and carriers docking and fusing with the target organelle for the delivery of cargo. Membrane tethers and small guanosine triphosphatases (GTPases) mediate the docking of transport vesicles/carriers to enhance the efficiency of the subsequent SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-mediated fusion event with the target membrane bilayer. Different classes of membrane tethers and their specific intracellular location throughout the endomembrane system are now well defined. Recent biochemical and structural studies have led to a deeper understanding of the mechanism by which membrane tethers mediate docking of membrane carriers as well as an appreciation of the role of tethers in coordinating the correct SNARE complex and in regulating the organization of membrane compartments. This review will summarize the properties and roles of membrane tethers of both secretory and endocytic systems.
Collapse
Affiliation(s)
- Pei Zhi Cheryl Chia
- National Institute of Dental and Craniofacial Research, National Institutes of Health30 Convent Drive, Bethesda, MD 20892-4340USA
| | - Paul A. Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute30 Flemington Road, The University of Melbourne, Victoria 3010Australia
| |
Collapse
|
38
|
Kobayashi H, Etoh K, Ohbayashi N, Fukuda M. Rab35 promotes the recruitment of Rab8, Rab13 and Rab36 to recycling endosomes through MICAL-L1 during neurite outgrowth. Biol Open 2014; 3:803-14. [PMID: 25086062 PMCID: PMC4163657 DOI: 10.1242/bio.20148771] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small GTPase Rab35 is an important molecular switch for endocytic recycling that regulates various cellular processes, including cytokinesis, cell migration, and neurite outgrowth. We previously showed that active Rab35 promotes nerve growth factor (NGF)-induced neurite outgrowth of PC12 cells by recruiting MICAL-L1, a multiple Rab-binding protein, to Arf6-positive recycling endosomes. However, the physiological significance of the multiple Rab-binding ability of MICAL-L1 during neurite outgrowth remained completely unknown. Here we report that Rab35 and MICAL-L1 promote the recruitment of Rab8, Rab13, and Rab36 to Arf6-positive recycling endosomes during neurite outgrowth. We found that Rab35 functions as a master Rab that determines the intracellular localization of MICAL-L1, which in turn functions as a scaffold for Rab8, Rab13, and Rab36. We further showed by functional ablation experiments that each of these downstream Rabs regulates neurite outgrowth in a non-redundant manner downstream of Rab35 and MICAL-L1, e.g. by showing that knockdown of Rab36 inhibited recruitment of Rab36-specific effector JIP4 to Arf6-positive recycling endosomes, and caused inhibition of neurite outgrowth without affecting accumulation of Rab8 and Rab13 in the same Arf6-positive area. Our findings suggest the existence of a novel mechanism that recruits multiple Rab proteins at the Arf6-positive compartment by MICAL-L1.
Collapse
Affiliation(s)
- Hotaka Kobayashi
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kan Etoh
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Norihiko Ohbayashi
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
39
|
Ailion M, Hannemann M, Dalton S, Pappas A, Watanabe S, Hegermann J, Liu Q, Han HF, Gu M, Goulding MQ, Sasidharan N, Schuske K, Hullett P, Eimer S, Jorgensen EM. Two Rab2 interactors regulate dense-core vesicle maturation. Neuron 2014; 82:167-80. [PMID: 24698274 DOI: 10.1016/j.neuron.2014.02.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2014] [Indexed: 12/14/2022]
Abstract
Peptide neuromodulators are released from a unique organelle: the dense-core vesicle. Dense-core vesicles are generated at the trans-Golgi and then sort cargo during maturation before being secreted. To identify proteins that act in this pathway, we performed a genetic screen in Caenorhabditis elegans for mutants defective in dense-core vesicle function. We identified two conserved Rab2-binding proteins: RUND-1, a RUN domain protein, and CCCP-1, a coiled-coil protein. RUND-1 and CCCP-1 colocalize with RAB-2 at the Golgi, and rab-2, rund-1, and cccp-1 mutants have similar defects in sorting soluble and transmembrane dense-core vesicle cargos. RUND-1 also interacts with the Rab2 GAP protein TBC-8 and the BAR domain protein RIC-19, a RAB-2 effector. In summary, a pathway of conserved proteins controls the maturation of dense-core vesicles at the trans-Golgi network.
Collapse
Affiliation(s)
- Michael Ailion
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Department of Biochemistry, University of Washington, Seattle WA, 98195, USA.
| | - Mandy Hannemann
- European Neuroscience Institute, 37077 Göttingen, Germany; International Max Planck Research School Molecular Biology, 37077 Göttingen, Germany
| | - Susan Dalton
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Andrea Pappas
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Shigeki Watanabe
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Jan Hegermann
- European Neuroscience Institute, 37077 Göttingen, Germany; DFG research Center for Molecular Physiology of the Brain (CMPB), 37077 Göttingen, Germany
| | - Qiang Liu
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Hsiao-Fen Han
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Mingyu Gu
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Morgan Q Goulding
- Department of Biochemistry, University of Washington, Seattle WA, 98195, USA
| | | | - Kim Schuske
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Patrick Hullett
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Stefan Eimer
- European Neuroscience Institute, 37077 Göttingen, Germany; DFG research Center for Molecular Physiology of the Brain (CMPB), 37077 Göttingen, Germany
| | - Erik M Jorgensen
- Howard Hughes Medical Institute, Department of Biology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
40
|
Wang S, Ma Z, Xu X, Wang Z, Sun L, Zhou Y, Lin X, Hong W, Wang T. A role of Rab29 in the integrity of the trans-Golgi network and retrograde trafficking of mannose-6-phosphate receptor. PLoS One 2014; 9:e96242. [PMID: 24788816 PMCID: PMC4008501 DOI: 10.1371/journal.pone.0096242] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 04/07/2014] [Indexed: 11/18/2022] Open
Abstract
Rab29 (also referred as Rab7L1) is a novel Rab protein, and is recently demonstrated to regulate phagocytosis and traffic from the Golgi to the lysosome. However, its roles in membrane trafficking have not been investigated extensively. Our results in this study revealed that Rab29 is associated with the trans-Golgi network (TGN), and is essential for maintaining the integrity of the TGN, because inhibition of the activity of Rab29 or depletion of Rab29 resulted in fragmentation of the TGN marked by TGN46. Expression of the dominant negative form Rab29T21N or shRNA-Rab29 also altered the distribution of mannose-6-phosphate receptor (M6PR), and interrupted the retrograde trafficking of M6PR through monitoring the endocytosis of CD8-tagged calcium dependent M6PR (cdM6PR) or calcium independent M6PR (ciM6PR), but without significant effects on the anterograde trafficking of vesicular stomatitis virus G protein (VSV-G). Our results suggest that Rab29 is essential for the integrity of the TGN and participates in the retrograde trafficking of M6PRs.
Collapse
Affiliation(s)
- Shicong Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Zexu Ma
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiaohui Xu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Zhen Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Lixiang Sun
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Yunhe Zhou
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiaosi Lin
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Wanjin Hong
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Tuanlao Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
41
|
Lu L, Hong W. From endosomes to the trans-Golgi network. Semin Cell Dev Biol 2014; 31:30-9. [PMID: 24769370 DOI: 10.1016/j.semcdb.2014.04.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 12/11/2022]
Abstract
The retrograde trafficking from endosomes to the trans-Golgi network (TGN) is one of the major endocytic pathways to divert proteins and lipids away from lysosomal degradation. Retrograde transported cargos enter the TGN via two itineraries from either the early endosome/recycling endosome or the late endosome and involve various machinery components such as retromer, sorting nexins, clathrin, small GTPases, tethering factors and SNAREs. Recently, the pathway has been recognized for its role in signal transduction, physiology and pathogenesis of human diseases.
Collapse
Affiliation(s)
- Lei Lu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; School of Pharmaceutical Sciences, Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
42
|
Rab6a/a' are important Golgi regulators of pro-inflammatory TNF secretion in macrophages. PLoS One 2013; 8:e57034. [PMID: 23437303 PMCID: PMC3578815 DOI: 10.1371/journal.pone.0057034] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/16/2013] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS)-activated macrophages secrete pro-inflammatory cytokines, including tumor necrosis factor (TNF) to elicit innate immune responses. Secretion of these cytokines is also a major contributing factor in chronic inflammatory disease. In previous studies we have begun to elucidate the pathways and molecules that mediate the intracellular trafficking and secretion of TNF. Rab6a and Rab6a' (collectively Rab6) are trans-Golgi-localized GTPases known for roles in maintaining Golgi structure and Golgi-associated trafficking. We found that induction of TNF secretion by LPS promoted the selective increase of Rab6 expression. Depletion of Rab6 (via siRNA and shRNA) resulted in reorganization of the Golgi ribbon into more compact structures that at the resolution of electron microcopy consisted of elongated Golgi stacks that likely arose from fusion of smaller Golgi elements. Concomitantly, the delivery of TNF to the cell surface and subsequent release into the media was reduced. Dominant negative mutants of Rab6 had similar effects in disrupting TNF secretion. In live cells, Rab6-GFP were localized on trans-Golgi network (TGN)-derived tubular carriers demarked by the golgin p230. Rab6 depletion and inactive mutants altered carrier egress and partially reduced p230 membrane association. Our results show that Rab6 acts on TNF trafficking at the level of TGN exit in tubular carriers and our findings suggest Rab6 may stabilize p230 on the tubules to facilitate TNF transport. Both Rab6 isoforms are needed in macrophages for Golgi stack organization and for the efficient post-Golgi transport of TNF. This work provides new insights into Rab6 function and into the role of the Golgi complex in cytokine secretion in inflammatory macrophages.
Collapse
|
43
|
Miller VJ, Sharma P, Kudlyk TA, Frost L, Rofe AP, Watson IJ, Duden R, Lowe M, Lupashin VV, Ungar D. Molecular insights into vesicle tethering at the Golgi by the conserved oligomeric Golgi (COG) complex and the golgin TATA element modulatory factor (TMF). J Biol Chem 2012; 288:4229-40. [PMID: 23239882 DOI: 10.1074/jbc.m112.426767] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein sorting between eukaryotic compartments requires vesicular transport, wherein tethering provides the first contact between vesicle and target membranes. Here we map and start to functionally analyze the interaction network of the conserved oligomeric Golgi (COG) complex that mediates retrograde tethering at the Golgi. The interactions of COG subunits with members of transport factor families assign the individual subunits as specific interaction hubs. Functional analysis of selected interactions suggests a mechanistic tethering model. We find that the COG complex interacts with two different Rabs in addition to each end of the golgin "TATA element modulatory factor" (TMF). This allows COG to potentially bridge the distance between the distal end of the golgin and the target membrane thereby promoting tighter docking. Concurrently we show that the central portion of TMF can bind to Golgi membranes that are liberated of their COPI cover. This latter interaction could serve to bring vesicle and target membranes into close apposition prior to fusion. A target selection mechanism, in which a hetero-oligomeric tethering factor organizes Rabs and coiled transport factors to enable protein sorting specificity, could be applicable to vesicle targeting throughout eukaryotic cells.
Collapse
Affiliation(s)
- Victoria J Miller
- Department of Biology, University of York, York, YO10 5DD, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Nottingham RM, Pusapati GV, Ganley IG, Barr FA, Lambright DG, Pfeffer SR. RUTBC2 protein, a Rab9A effector and GTPase-activating protein for Rab36. J Biol Chem 2012; 287:22740-8. [PMID: 22637480 DOI: 10.1074/jbc.m112.362558] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rab GTPases regulate vesicle budding, motility, docking, and fusion. In cells, their cycling between active, GTP-bound states and inactive, GDP-bound states is regulated by the action of opposing enzymes called guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). The substrates for most RabGAPs are unknown, and the potential for cross-talk between different membrane trafficking pathways remains uncharted territory. Rab9A and its effectors regulate recycling of mannose 6-phosphate receptors from late endosomes to the trans Golgi network. We show here that RUTBC2 is a TBC domain-containing protein that binds to Rab9A specifically both in vitro and in cultured cells but is not a GAP for Rab9A. Biochemical screening of Rab protein substrates for RUTBC2 revealed highest GAP activity toward Rab34 and Rab36. In cells, membrane-associated RUTBC2 co-localizes with Rab36, and expression of wild type RUTBC2, but not the catalytically inactive, RUTBC2 R829A mutant, decreases the amount of membrane-associated Rab36 protein. These data show that RUTBC2 can act as a Rab36 GAP in cells and suggest that RUTBC2 links Rab9A function to Rab36 function in the endosomal system.
Collapse
Affiliation(s)
- Ryan M Nottingham
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Members of the Rab or ARF/Sar branches of the Ras GTPase superfamily regulate almost every step of intracellular membrane traffic. A rapidly growing body of evidence indicates that these GTPases do not act as lone agents but are networked to one another through a variety of mechanisms to coordinate the individual events of one stage of transport and to link together the different stages of an entire transport pathway. These mechanisms include guanine nucleotide exchange factor (GEF) cascades, GTPase-activating protein (GAP) cascades, effectors that bind to multiple GTPases, and positive-feedback loops generated by exchange factor-effector interactions. Together these mechanisms can lead to an ordered series of transitions from one GTPase to the next. As each GTPase recruits a unique set of effectors, these transitions help to define changes in the functionality of the membrane compartments with which they are associated.
Collapse
Affiliation(s)
- Emi Mizuno-Yamasaki
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan.
| | | | | |
Collapse
|
46
|
Intrinsic tethering activity of endosomal Rab proteins. Nat Struct Mol Biol 2011; 19:40-7. [PMID: 22157956 PMCID: PMC3252480 DOI: 10.1038/nsmb.2162] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 09/22/2011] [Indexed: 12/24/2022]
Abstract
Rab small G-proteins control membrane trafficking events required for a multitude of processes including secretion, lipid metabolism, antigen presentation, and growth factor signaling. Rabs recruit effectors that mediate diverse functions including vesicle tethering and fusion. However, many mechanistic questions about Rab-regulated vesicle tethering are unresolved. Using chemically defined reaction systems we discovered that Vps21, a Saccharomyces cerevisiae ortholog of mammalian endosomal Rab5, functions in trans with itself and with at least two other endosomal Rabs to directly mediate GTP-dependent tethering. Vps21-mediated tethering was stringently and reversibly regulated by an upstream activator, Vps9, and an inhibitor, Gyp1, which were sufficient to drive dynamic cycles of tethering and de-tethering. These experiments reveal an unexpected mode of tethering by endocytic Rabs. In our working model, the intrinsic tethering capacity Vps21 operates in concert with conventional effectors and SNAREs to drive efficient docking and fusion.
Collapse
|
47
|
Lin YC, Chiang TC, Liu YT, Tsai YT, Jang LT, Lee FJS. ARL4A acts with GCC185 to modulate Golgi complex organization. J Cell Sci 2011; 124:4014-26. [PMID: 22159419 DOI: 10.1242/jcs.086892] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
ADP-ribosylation factor-like protein 4A (ARL4A) is a developmentally regulated member of the ARF/ARL GTPase family. The primary structure of ARL4A is very similar to that of other ARF/ARL molecules, but its function remains unclear. The trans-Golgi network golgin GCC185 is required for maintenance of Golgi structure and distinct endosome-to-Golgi transport. We show here that GCC185 acts as a new effector for ARL4 to modulate Golgi organization. ARL4A directly interacts with GCC185 in a GTP-dependent manner. Sub-coiled-coil regions of the CC2 domain of GCC185 are required for the interaction between GCC185 and ARL4A. Depletion of ARL4A reproduces the GCC185-depleted phenotype, causing fragmentation of the Golgi compartment and defects in endosome-to-Golgi transport. GCC185 and ARL4A localize to the Golgi independently of each other. Deletion of the ARL4A-interacting region of GCC185 results in inability to maintain Golgi structure. Depletion of ARL4A impairs the interaction between GCC185 and cytoplasmic linker-associated proteins 1 and 2 (CLASP1 and CLASP2, hereafter CLASPs) in vivo, and abolishes the GCC185-mediated Golgi recruitment of these CLASPs, which is crucial for the maintenance of Golgi structure. In summary, we suggest that ARL4A alters the integrity of the Golgi structure by facilitating the interaction of GCC185 with CLASPs.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
48
|
Kümmel D, Reinisch KM. Structure of Golgi transport proteins. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a007609. [PMID: 21813399 DOI: 10.1101/cshperspect.a007609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The function of the Golgi has long been recognized to critically depend on vesicular transport from, to, and within its cisternae, involving constant membrane fission and fusion. These processes are mediated by Arf GTPases and coat proteins, and Rabs, tethers and SNARE proteins, respectively. In this article, we describe structural studies of Golgi coats and tethers and their interactions with SNAREs and GTPases as well as insights regarding membrane traffic processes that these have provided.
Collapse
Affiliation(s)
- Daniel Kümmel
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | |
Collapse
|
49
|
Affiliation(s)
| | - Adam D. Linstedt
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15232;
| |
Collapse
|
50
|
Abstract
Bacteria and eukaryotic cells contain geometry-sensing tools in their cytosol: protein motifs or domains that recognize the curvature, concave or convex, deep or shallow, of lipid membranes. These sensors contrast with classical lipid-binding domains by their extended structure and, sometimes, counterintuitive chemistry. Among the sensors are long amphipathic helices, such as the ALPS motif and the N-terminal region of α-synuclein, whose apparent "design defects" translate into a remarkable ability to specifically adsorb to the surface of small vesicles. Fundamental differences in the lipid composition of membranes of the early and late secretory pathways probably explain why some sensors use mostly electrostatics whereas others take advantage of the hydrophobic effect. Membrane curvature sensors help to organize very diverse reactions, such as lipid transfer between membranes, the tethering of vesicles at the Golgi apparatus, and the assembly-disassembly cycle of protein coats.
Collapse
Affiliation(s)
- Bruno Antonny
- Université de Nice-Sophia Antipolis and Centre National de la Recheche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France.
| |
Collapse
|