1
|
Dwivedi A, Scalsky RJ, Harris DG, Stabler TC, Shrestha B, Joshi S, Gandhi C, Munro JB, Ifeonu OO, Ouedraogo A, Tiono AB, Coulibaly D, Ouattara A, Richie TL, Sim BKL, Plowe CV, Lyke KE, Takala-Harrison S, Hoffman SL, Thera MA, Sirima SB, Laurens MB, Silva JC. Protective targets of PfSPZ vaccines identified from whole-genome sieve analysis of isolates from malaria vaccine efficacy trials in West Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.04.25323352. [PMID: 40093207 PMCID: PMC11908318 DOI: 10.1101/2025.03.04.25323352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Identification of antigens targeted by a protective response is a central quest in malaria vaccinology. Whole-genome sieve analysis (SAWG) in samples collected from placebo-controlled field trials of Plasmodium falciparum (Pf) sporozoite (SPZ) vaccines may enable identification of Pf pre-erythrocytic antigens. We applied SAWG to genomic data generated from Pf isolates collected during two field trials measuring the efficacy, in malaria-exposed African adults, of two PfSPZ vaccines. These randomized, double-blind, placebo-controlled trials were conducted in regions of Mali and Burkina Faso characterized by high seasonal transmission, where parasite genetic diversity is high. Genomic sites in which the vaccine allelic state was significantly underrepresented among breakthrough infections in vaccinees relative to placebo recipients were termed "target sites". Protein-coding loci containing target sites that changed amino acids were termed "target loci". The SAWG conducted on clinical trial samples from the Burkina Faso and Mali trials identified 138 and 80 single-copy protein-coding target loci in the Burkinabe and Malian data sets, respectively, with twelve common to both, a number significantly higher than expected (E = 3.9; 99%CI = [0, 9]). Among these was the thrombospondin-related anonymous protein locus, which encodes PfSSP2|TRAP, one of the most abundant and well-characterized pre-erythrocytic stage antigen as well as other genes encoding membrane-associated proteins of unknown function. These results identify SAWG as a potentially powerful tool for identifying protective vaccine antigens in recombining pathogens with large genome size and reveals potential new protective Pf antigens.
Collapse
Affiliation(s)
- Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Ryan J. Scalsky
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - David G. Harris
- Department of Computer Science, University of Maryland College Park; College Park, MD 20742, USA
| | | | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Chakshu Gandhi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - James B. Munro
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Olukemi O. Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Alfred B. Tiono
- Groupe de Recherche Action en Santé; Ouagadougou, Burkina Faso
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | - Amed Ouattara
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | | | - Christopher V. Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Shannon Takala-Harrison
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (NOVA); 1349-008 Lisboa, Portugal
| |
Collapse
|
2
|
de Voogt WS, Frunt R, Leandro RM, Triesscheijn CS, Monica B, Paspali I, Tielemans M, François JJJM, Seinen CW, de Jong OG, Kooijmans SAA. EV-Elute: A universal platform for the enrichment of functional surface marker-defined extracellular vesicle subpopulations. J Extracell Vesicles 2024; 13:e70017. [PMID: 39692115 DOI: 10.1002/jev2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 12/19/2024] Open
Abstract
Intercellular communication via extracellular vesicles (EVs) has been identified as a vital component of a steadily expanding number of physiological and pathological processes. To accommodate these roles, EVs have highly heterogeneous molecular compositions. Given that surface molecules on EVs determine their interactions with their environment, EV functionality likely differs between subpopulations with varying surface compositions. However, it has been technically challenging to examine such functional heterogeneity due to a lack of non-destructive methods to separate EV subpopulations based on their surface markers. Here, we used the Design-of-Experiments (DoE) methodology to optimize a protocol, which we name 'EV-Elute', to elute intact EVs from commercially available Protein G-coated magnetic beads. We captured EVs from various cell types on these beads using antibodies against CD9, CD63, CD81 and a custom-made protein binding phosphatidylserine (PS). When applying EV-Elute, over 70% of bound EVs could be recovered from the beads in a pH- and incubation-time-dependent fashion. EV subpopulations showed intact integrity by electron microscopy and Proteinase K protection assays and showed uptake patterns similar to whole EV isolates in co-cultures of peripheral blood mononuclear cells (PBMCs) and endothelial cells. However, in Cas9/sgRNA delivery assays, CD63+ EVs showed a lower capacity to functionally deliver cargo as compared to CD9+, CD81+ and PS+ EVs. Taken together, we developed a novel, easy-to-use platform to isolate and functionally compare surface marker-defined EV subpopulations. This platform does not require specialized equipment or reagents and is universally applicable to any capturing antibody and EV source. Hence, EV-Elute can open new opportunities to study EV functionality at the subpopulation level.
Collapse
Affiliation(s)
| | - Rowan Frunt
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Raul M Leandro
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Bella Monica
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ioanna Paspali
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark Tielemans
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Cor W Seinen
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Sander A A Kooijmans
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
- Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Metabolic Diseases, Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Castello-Serrano I, Heberle FA, Diaz-Rohrer B, Ippolito R, Shurer CR, Lujan P, Campelo F, Levental KR, Levental I. Partitioning to ordered membrane domains regulates the kinetics of secretory traffic. eLife 2024; 12:RP89306. [PMID: 38837189 PMCID: PMC11152573 DOI: 10.7554/elife.89306] [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] [Indexed: 06/06/2024] Open
Abstract
The organelles of eukaryotic cells maintain distinct protein and lipid compositions required for their specific functions. The mechanisms by which many of these components are sorted to their specific locations remain unknown. While some motifs mediating subcellular protein localization have been identified, many membrane proteins and most membrane lipids lack known sorting determinants. A putative mechanism for sorting of membrane components is based on membrane domains known as lipid rafts, which are laterally segregated nanoscopic assemblies of specific lipids and proteins. To assess the role of such domains in the secretory pathway, we applied a robust tool for synchronized secretory protein traffic (RUSH, Retention Using Selective Hooks) to protein constructs with defined affinity for raft phases. These constructs consist solely of single-pass transmembrane domains (TMDs) and, lacking other sorting determinants, constitute probes for membrane domain-mediated trafficking. We find that while raft affinity can be sufficient for steady-state PM localization, it is not sufficient for rapid exit from the endoplasmic reticulum (ER), which is instead mediated by a short cytosolic peptide motif. In contrast, we find that Golgi exit kinetics are highly dependent on raft affinity, with raft preferring probes exiting the Golgi ~2.5-fold faster than probes with minimal raft affinity. We rationalize these observations with a kinetic model of secretory trafficking, wherein Golgi export can be facilitated by protein association with raft domains. These observations support a role for raft-like membrane domains in the secretory pathway and establish an experimental paradigm for dissecting its underlying machinery.
Collapse
Affiliation(s)
- Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | | | | | - Rossana Ippolito
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Carolyn R Shurer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Pablo Lujan
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Felix Campelo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
4
|
Castello-Serrano I, Heberle FA, Diaz-Rohrer B, Ippolito R, Shurer CR, Lujan P, Campelo F, Levental KR, Levental I. Partitioning to ordered membrane domains regulates the kinetics of secretory traffic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.18.537395. [PMID: 37131599 PMCID: PMC10153169 DOI: 10.1101/2023.04.18.537395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The organelles of eukaryotic cells maintain distinct protein and lipid compositions required for their specific functions. The mechanisms by which many of these components are sorted to their specific locations remain unknown. While some motifs mediating subcellular protein localization have been identified, many membrane proteins and most membrane lipids lack known sorting determinants. A putative mechanism for sorting of membrane components is based on membrane domains known as lipid rafts, which are laterally segregated nanoscopic assemblies of specific lipids and proteins. To assess the role of such domains in the secretory pathway, we applied a robust tool for synchronized secretory protein traffic (RUSH, Retention Using Selective Hooks) to protein constructs with defined affinity for raft phases. These constructs consist solely of single-pass transmembrane domains (TMDs) and, lacking other sorting determinants, constitute probes for membrane domain-mediated trafficking. We find that while raft affinity can be sufficient for steady-state PM localization, it is not sufficient for rapid exit from the endoplasmic reticulum (ER), which is instead mediated by a short cytosolic peptide motif. In contrast, we find that Golgi exit kinetics are highly dependent on raft affinity, with raft preferring probes exiting Golgi ~2.5-fold faster than probes with minimal raft affinity. We rationalize these observations with a kinetic model of secretory trafficking, wherein Golgi export can be facilitated by protein association with raft domains. These observations support a role for raft-like membrane domains in the secretory pathway and establish an experimental paradigm for dissecting its underlying machinery.
Collapse
|
5
|
Xu H, Chang F, Jain S, Heller BA, Han X, Liu Y, Edwards RH. SNX5 targets a monoamine transporter to the TGN for assembly into dense core vesicles by AP-3. J Cell Biol 2022; 221:e202106083. [PMID: 35426896 PMCID: PMC9016777 DOI: 10.1083/jcb.202106083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/06/2021] [Accepted: 02/16/2022] [Indexed: 11/22/2022] Open
Abstract
The time course of signaling by peptide hormones, neural peptides, and other neuromodulators depends on their storage inside dense core vesicles (DCVs). Adaptor protein 3 (AP-3) assembles the membrane proteins that confer regulated release of DCVs and is thought to promote their trafficking from endosomes directly to maturing DCVs. We now find that regulated monoamine release from DCVs requires sorting nexin 5 (SNX5). Loss of SNX5 disrupts trafficking of the vesicular monoamine transporter (VMAT) to DCVs. The mechanism involves a role for SNX5 in retrograde transport of VMAT from endosomes to the TGN. However, this role for SNX5 conflicts with the proposed function of AP-3 in trafficking from endosomes directly to DCVs. We now identify a transient role for AP-3 at the TGN, where it associates with DCV cargo. Thus, retrograde transport from endosomes by SNX5 enables DCV assembly at the TGN by AP-3, resolving the apparent antagonism. A novel role for AP-3 at the TGN has implications for other organelles that also depend on this adaptor.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Fei Chang
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Shweta Jain
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Bradley Austin Heller
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Xu Han
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yongjian Liu
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Departments of Pharmacology and Biological Chemistry, University of Pittsburgh, Pittsburgh, PA
| | - Robert H. Edwards
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
6
|
Shin J, Nile A, Oh JW. Role of adaptin protein complexes in intracellular trafficking and their impact on diseases. Bioengineered 2021; 12:8259-8278. [PMID: 34565296 PMCID: PMC8806629 DOI: 10.1080/21655979.2021.1982846] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
Adaptin proteins (APs) play a crucial role in intracellular cell trafficking. The 'classical' role of APs is carried out by AP1‒3, which bind to clathrin, cargo, and accessory proteins. Accordingly, AP1-3 are crucial for both vesicle formation and sorting. All APs consist of four subunits that are indispensable for their functions. In fact, based on studies using cells, model organism knockdown/knock-out, and human variants, each subunit plays crucial roles and contributes to the specificity of each AP. These studies also revealed that the sorting and intracellular trafficking function of AP can exert varying effects on pathology by controlling features such as cell development, signal transduction related to the apoptosis and proliferation pathways in cancer cells, organelle integrity, receptor presentation, and viral infection. Although the roles and functions of AP1‒3 are relatively well studied, the functions of the less abundant and more recently identified APs, AP4 and AP5, are still to be investigated. Further studies on these APs may enable a better understanding and targeting of specific diseases.APs known or suggested locations and functions.
Collapse
Affiliation(s)
- Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| | - Arti Nile
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Viral Interactions with Adaptor-Protein Complexes: A Ubiquitous Trait among Viral Species. Int J Mol Sci 2021; 22:ijms22105274. [PMID: 34067854 PMCID: PMC8156722 DOI: 10.3390/ijms22105274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022] Open
Abstract
Numerous viruses hijack cellular protein trafficking pathways to mediate cell entry or to rearrange membrane structures thereby promoting viral replication and antagonizing the immune response. Adaptor protein complexes (AP), which mediate protein sorting in endocytic and secretory transport pathways, are one of the conserved viral targets with many viruses possessing AP-interacting motifs. We present here different mechanisms of viral interference with AP complexes and the functional consequences that allow for efficient viral propagation and evasion of host immune defense. The ubiquity of this phenomenon is evidenced by the fact that there are representatives for AP interference in all major viral families, covered in this review. The best described examples are interactions of human immunodeficiency virus and human herpesviruses with AP complexes. Several other viruses, like Ebola, Nipah, and SARS-CoV-2, are pointed out as high priority disease-causative agents supporting the need for deeper understanding of virus-AP interplay which can be exploited in the design of novel antiviral therapies.
Collapse
|
8
|
Lavie M, Linna L, Moustafa RI, Belouzard S, Fukasawa M, Dubuisson J. Role of the cytosolic domain of occludin in trafficking and hepatitis C virus infection. Traffic 2019; 20:753-773. [PMID: 31328852 DOI: 10.1111/tra.12680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022]
Abstract
The role of the tight-junction (TJ) protein occludin (OCLN) in hepatitis C virus (HCV) entry remains elusive. Here, we investigated the OCLN C-terminal cytosolic domain in HCV infection. We expressed a series of C-terminal deletion mutants in Huh-7 cells KO for OCLN and characterized their functionality in HCV infection and trafficking. Deleting the OCLN cytosolic domain led to protein instability and intracellular retention. The first 15 residues (OCLN-C15 mutant) of the cytosolic domain were sufficient for OCLN stability, but led to its accumulation in the trans-Golgi network (TGN) due to a deficient cell surface export after synthesis. In contrast, the OCLN-C18 mutant, containing the first 18 residues of the cytosolic domain, was expressed at the cell surface and could mediate HCV infection. Point mutations in the context of C18 showed that I279 and W281 are crucial residues for cell surface expression of OCLN-C18. However, in the context of full-length OCLN, mutation of these residues only partially affected infection and cell surface localization. Importantly, the characterization of OCLN-C18 in human-polarized hepatocytes revealed a defect in its TJ localization without affecting HCV infection. These data suggest that TJ localization of OCLN is not a prerequisite for HCV infection in polarized hepatocytes.
Collapse
Affiliation(s)
- Muriel Lavie
- Universite de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Lydia Linna
- Universite de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Rehab I Moustafa
- Universite de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Lille, France.,Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Division, National Research Center, Cairo, Egypt
| | - Sandrine Belouzard
- Universite de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Masayoshi Fukasawa
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Jean Dubuisson
- Universite de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
9
|
Ward C, Maselko M, Lupfer C, Prescott M, Pastey MK. Interaction of the Human Respiratory Syncytial Virus matrix protein with cellular adaptor protein complex 3 plays a critical role in trafficking. PLoS One 2017; 12:e0184629. [PMID: 29028839 PMCID: PMC5640227 DOI: 10.1371/journal.pone.0184629] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/28/2017] [Indexed: 01/03/2023] Open
Abstract
Human Respiratory Syncytial Virus (HRSV) is a leading cause of bronchopneumonia in infants and the elderly. To date, knowledge of viral and host protein interactions within HRSV is limited and are critical areas of research. Here, we show that HRSV Matrix (M) protein interacts with the cellular adaptor protein complex 3 specifically via its medium subunit (AP-3Mu3A). This novel protein-protein interaction was first detected via yeast-two hybrid screen and was further confirmed in a mammalian system by immunofluorescence colocalization and co-immunoprecipitation. This novel interaction is further substantiated by the presence of a known tyrosine-based adaptor protein MU subunit sorting signal sequence, YXXФ: where Ф is a bulky hydrophobic residue, which is conserved across the related RSV M proteins. Analysis of point-mutated HRSV M derivatives indicated that AP-3Mu3A- mediated trafficking is contingent on the presence of the tyrosine residue within the YXXL sorting sequence at amino acids 197–200 of the M protein. AP-3Mu3A is up regulated at 24 hours post-infection in infected cells versus mock-infected HEp2 cells. Together, our data suggests that the AP-3 complex plays a critical role in the trafficking of HRSV proteins specifically matrix in epithelial cells. The results of this study add new insights and targets that may lead to the development of potential antivirals and attenuating mutations suitable for candidate vaccines in the future.
Collapse
Affiliation(s)
- Casey Ward
- Department of Veterinary Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Maciej Maselko
- Department of Veterinary Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Christopher Lupfer
- Department of Veterinary Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Meagan Prescott
- Department of Veterinary Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Manoj K. Pastey
- Department of Veterinary Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
10
|
Caceres PS, Benedicto I, Lehmann GL, Rodriguez-Boulan EJ. Directional Fluid Transport across Organ-Blood Barriers: Physiology and Cell Biology. Cold Spring Harb Perspect Biol 2017; 9:a027847. [PMID: 28003183 PMCID: PMC5334253 DOI: 10.1101/cshperspect.a027847] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.
Collapse
Affiliation(s)
- Paulo S Caceres
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Ignacio Benedicto
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Guillermo L Lehmann
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Enrique J Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
11
|
Parmar HB, Duncan R. A novel tribasic Golgi export signal directs cargo protein interaction with activated Rab11 and AP-1-dependent Golgi-plasma membrane trafficking. Mol Biol Cell 2016; 27:1320-31. [PMID: 26941330 PMCID: PMC4831885 DOI: 10.1091/mbc.e15-12-0845] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/23/2016] [Indexed: 12/30/2022] Open
Abstract
A novel sorting motif present in the reovirus p14 fusion–associated small transmembrane protein directs interaction with GTP-Rab11 at the TGN and sorting into AP-1–coated vesicles for trafficking to the plasma membrane. This is the first example of cargo protein interaction with activated Rab11 mediating anterograde trafficking from the TGN. The reovirus fusion–associated small transmembrane (FAST) proteins comprise a unique family of viral membrane fusion proteins dedicated to inducing cell–cell fusion. We recently reported that a polybasic motif (PBM) in the cytosolic tail of reptilian reovirus p14 FAST protein functions as a novel tribasic Golgi export signal. Using coimmunoprecipitation and fluorescence resonance energy transfer (FRET) assays, we now show the PBM directs interaction of p14 with GTP-Rab11. Overexpression of dominant-negative Rab11 and RNA interference knockdown of endogenous Rab11 inhibited p14 plasma membrane trafficking and resulted in p14 accumulation in the Golgi complex. This is the first example of Golgi export to the plasma membrane that is dependent on the interaction of membrane protein cargo with activated Rab11. RNA interference and immunofluorescence microscopy further revealed that p14 Golgi export is dependent on AP-1 (but not AP-3 or AP-4) and that Rab11 and AP-1 both colocalize with p14 at the TGN. Together these results imply the PBM mediates interactions of p14 with activated Rab11 at the TGN, resulting in p14 sorting into AP1-coated vesicles for anterograde TGN–plasma membrane transport.
Collapse
Affiliation(s)
- Hirendrasinh B Parmar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Roy Duncan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
12
|
Zhang M, Davis JE, Li C, Gao J, Huang W, Lambert NA, Terry AV, Wu G. GGA3 Interacts with a G Protein-Coupled Receptor and Modulates Its Cell Surface Export. Mol Cell Biol 2016; 36:1152-63. [PMID: 26811329 PMCID: PMC4800796 DOI: 10.1128/mcb.00009-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/20/2016] [Indexed: 12/20/2022] Open
Abstract
Molecular mechanisms governing the anterograde trafficking of nascent G protein-coupled receptors (GPCRs) are poorly understood. Here, we have studied the regulation of cell surface transport of α2-adrenergic receptors (α2-ARs) by GGA3 (Golgi-localized, γ-adaptin ear domain homology, ADP ribosylation factor-binding protein 3), a multidomain clathrin adaptor protein that sorts cargo proteins at the trans-Golgi network (TGN) to the endosome/lysosome pathway. By using an inducible system, we demonstrated that GGA3 knockdown significantly inhibited the cell surface expression of newly synthesized α2B-AR without altering overall receptor synthesis and internalization. The receptors were arrested in the TGN. Furthermore, GGA3 knockdown attenuated α2B-AR-mediated signaling, including extracellular signal-regulated kinase 1/2 (ERK1/2) activation and cyclic AMP (cAMP) inhibition. More interestingly, GGA3 physically interacted with α2B-AR, and the interaction sites were identified as the triple Arg motif in the third intracellular loop of the receptor and the acidic motif EDWE in the VHS domain of GGA3. In contrast, α2A-AR did not interact with GGA3 and its cell surface export and signaling were not affected by GGA3 knockdown. These data reveal a novel function of GGA3 in export trafficking of a GPCR that is mediated via a specific interaction with the receptor.
Collapse
Affiliation(s)
- Maoxiang Zhang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jason E Davis
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Chunman Li
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jie Gao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Wei Huang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
13
|
Regulation of GPCR Anterograde Trafficking by Molecular Chaperones and Motifs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:289-305. [PMID: 26055064 DOI: 10.1016/bs.pmbts.2015.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
G protein-coupled receptors (GPCRs) make up a superfamily of integral membrane proteins that respond to a wide variety of extracellular stimuli, giving them an important role in cell function and survival. They have also proven to be valuable targets in the fight against various diseases. As such, GPCR signal regulation has received considerable attention over the last few decades. With the amplitude of signaling being determined in large part by receptor density at the plasma membrane, several endogenous mechanisms for modulating GPCR expression at the cell surface have come to light. It has been shown that cell surface expression is determined by both exocytic and endocytic processes. However, the body of knowledge surrounding GPCR trafficking from the endoplasmic reticulum to the plasma membrane, commonly known as anterograde trafficking, has considerable room for growth. We focus here on the current paradigms of anterograde GPCR trafficking. We will discuss the regulatory role of both the general and "nonclassical private" chaperone systems in GPCR trafficking as well as conserved motifs that serve as modulators of GPCR export from the endoplasmic reticulum and Golgi apparatus. Together, these topics summarize some of the known mechanisms by which the cell regulates anterograde GPCR trafficking.
Collapse
|
14
|
Wu G, Davis JE, Zhang M. Regulation of α2B-Adrenerigc Receptor Export Trafficking by Specific Motifs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:227-44. [PMID: 26055061 PMCID: PMC4827153 DOI: 10.1016/bs.pmbts.2015.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Intracellular trafficking and precise targeting to specific locations of G protein-coupled receptors (GPCRs) control the physiological functions of the receptors. Compared to the extensive efforts dedicated to understanding the events involved in the endocytic and recycling pathways, the molecular mechanisms underlying the transport of the GPCR superfamily from the endoplasmic reticulum (ER) through the Golgi to the plasma membrane are relatively less well defined. Over the past years, we have used α(2B)-adrenergic receptor (α(2B)-AR) as a model to define the factors that control GPCR export trafficking. In this chapter, we will review specific motifs identified to mediate the export of nascent α(2B)-AR from the ER and the Golgi and discuss the possible underlying mechanisms. As these motifs are highly conserved among GPCRs, they may provide common mechanisms for export trafficking of these receptors.
Collapse
Affiliation(s)
- Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA.
| | - Jason E Davis
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | - Maoxiang Zhang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
15
|
Myeong J, Kwak M, Hong C, Jeon JH, So I. Identification of a membrane-targeting domain of the transient receptor potential canonical (TRPC)4 channel unrelated to its formation of a tetrameric structure. J Biol Chem 2014; 289:34990-5002. [PMID: 25349210 DOI: 10.1074/jbc.m114.584649] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are Ca(2+)-permeable nonselective cation channels that are activated by a wide variety of stimuli, including G protein-coupled receptors (GPCRs). The TRPC4 channel is expressed in a punctate distribution in the membrane. To identify the regulating region of the channel trafficking to the membrane, we generated deletion mutants of the TRPC4 channel. We determined that when either region that was downstream of the 20 amino acids of the N terminus or the 700-730 amino acids was deleted, the mutants were retained in the endoplasmic reticulum. By coexpression of the wild-type TRPC4 with deletion mutants, we found that the 23-29 amino acids of the N terminus regulate a membrane trafficking. Additionally, by the fluorescence resonance energy transfer (FRET) method, we found that the regions downstream of the 99 amino acid region of the N terminus and upstream of the 730 amino acid region in the C terminus produce assembly of the TRPC4 tetramers. We inferred the candidate proteins that regulate or interact with the 23-29 domain of TRPC4.
Collapse
Affiliation(s)
- Jongyun Myeong
- From the Department of Physiology, Seoul National University College of Medicine
| | - Misun Kwak
- From the Department of Physiology, Seoul National University College of Medicine
| | - Chansik Hong
- From the Department of Physiology, Seoul National University College of Medicine
| | - Ju-Hong Jeon
- From the Department of Physiology, Seoul National University College of Medicine
| | - Insuk So
- From the Department of Physiology, Seoul National University College of Medicine
| |
Collapse
|
16
|
Yu S, Yehia G, Wang J, Stypulkowski E, Sakamori R, Jiang P, Hernandez-Enriquez B, Tran TS, Bonder EM, Guo W, Gao N. Global ablation of the mouse Rab11a gene impairs early embryogenesis and matrix metalloproteinase secretion. J Biol Chem 2014; 289:32030-32043. [PMID: 25271168 DOI: 10.1074/jbc.m113.538223] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rab11a has been conceived as a prominent regulatory component of the recycling endosome, which acts as a nexus in the endo- and exocytotic networks. The precise in vivo role of Rab11a in mouse embryonic development is unknown. We globally ablated Rab11a and examined the phenotypic and molecular outcomes in Rab11a(null) blastocysts and mouse embryonic fibroblasts. Using multiple trafficking assays and complementation analyses, we determined, among multiple important membrane-associated and soluble cargos, the critical contribution of Rab11a vesicular traffic to the secretion of multiple soluble MMPs. Rab11a(null) embryos were able to properly form normal blastocysts but died at peri-implantation stages. Our data suggest that Rab11a critically controls mouse blastocyst development and soluble matrix metalloproteinase secretion.
Collapse
Affiliation(s)
- Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ghassan Yehia
- Transgenic Core Facility, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Juanfei Wang
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Ewa Stypulkowski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ping Jiang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | | | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102,.
| |
Collapse
|
17
|
Fossati M, Colombo SF, Borgese N. A positive signal prevents secretory membrane cargo from recycling between the Golgi and the ER. EMBO J 2014; 33:2080-97. [PMID: 25063674 DOI: 10.15252/embj.201488367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The Golgi complex and ER are dynamically connected by anterograde and retrograde trafficking pathways. To what extent and by what mechanism outward-bound cargo proteins escape retrograde trafficking has been poorly investigated. Here, we analysed the behaviour of several membrane proteins at the ER/Golgi interface in live cells. When Golgi-to-plasma membrane transport was blocked, vesicular stomatitis virus glycoprotein (VSVG), which bears an ER export signal, accumulated in the Golgi, whereas an export signal-deleted version of VSVG attained a steady state determined by the balance of retrograde and anterograde traffic. A similar behaviour was displayed by EGF receptor and by a model tail-anchored protein, whose retrograde traffic was slowed by addition of VSVG's export signal. Retrograde trafficking was energy- and Rab6-dependent, and Rab6 inhibition accelerated signal-deleted VSVG's transport to the cell surface. Our results extend the dynamic bi-directional relationship between the Golgi and ER to include surface-directed proteins, uncover an unanticipated role for export signals at the Golgi complex, and identify recycling as a novel factor that regulates cargo transport out of the early secretory pathway.
Collapse
Affiliation(s)
- Matteo Fossati
- BIOMETRA Department, CNR Institute of Neuroscience, Università degli Studi di Milano, Milano, Italy
| | - Sara F Colombo
- BIOMETRA Department, CNR Institute of Neuroscience, Università degli Studi di Milano, Milano, Italy
| | - Nica Borgese
- BIOMETRA Department, CNR Institute of Neuroscience, Università degli Studi di Milano, Milano, Italy Department of Health Science, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
18
|
Rodriguez-Boulan E, Macara IG. Organization and execution of the epithelial polarity programme. Nat Rev Mol Cell Biol 2014; 15:225-42. [PMID: 24651541 DOI: 10.1038/nrm3775] [Citation(s) in RCA: 528] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epithelial cells require apical-basal plasma membrane polarity to carry out crucial vectorial transport functions and cytoplasmic polarity to generate different cell progenies for tissue morphogenesis. The establishment and maintenance of a polarized epithelial cell with apical, basolateral and ciliary surface domains is guided by an epithelial polarity programme (EPP) that is controlled by a network of protein and lipid regulators. The EPP is organized in response to extracellular cues and is executed through the establishment of an apical-basal axis, intercellular junctions, epithelial-specific cytoskeletal rearrangements and a polarized trafficking machinery. Recent studies have provided insight into the interactions of the EPP with the polarized trafficking machinery and how these regulate epithelial polarization and depolarization.
Collapse
Affiliation(s)
- Enrique Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Weill Cornell Medical College, 1300 York Avenue, LC-301 New York City, New York 10065, USA
| | - Ian G Macara
- Department of Cell & Developmental Biology, Vanderbilt University Medical Center, 465 21st Avenue South, U 3209 MRB III, Nashville Tennessee 37232, USA
| |
Collapse
|
19
|
Junking M, Sawasdee N, Duangtum N, Cheunsuchon B, Limjindaporn T, Yenchitsomanus PT. Role of adaptor proteins and clathrin in the trafficking of human kidney anion exchanger 1 (kAE1) to the cell surface. Traffic 2014; 15:788-802. [PMID: 24698155 DOI: 10.1111/tra.12172] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/30/2014] [Accepted: 03/30/2014] [Indexed: 12/31/2022]
Abstract
Kidney anion exchanger 1 (kAE1) plays an important role in acid-base homeostasis by mediating chloride/bicarbornate (Cl-/HCO3-) exchange at the basolateral membrane of α-intercalated cells in the distal nephron. Impaired intracellular trafficking of kAE1 caused by mutations of SLC4A1 encoding kAE1 results in kidney disease - distal renal tubular acidosis (dRTA). However, it is not known how the intracellular sorting and trafficking of kAE1 from trans-Golgi network (TGN) to the basolateral membrane occurs. Here, we studied the role of basolateral-related sorting proteins, including the mu1 subunit of adaptor protein (AP) complexes, clathrin and protein kinase D, on kAE1 trafficking in polarized and non-polarized kidney cells. By using RNA interference, co-immunoprecipitation, yellow fluorescent protein-based protein fragment complementation assays and immunofluorescence staining, we demonstrated that AP-1 mu1A, AP-3 mu1, AP-4 mu1 and clathrin (but not AP-1 mu1B, PKD1 or PKD2) play crucial roles in intracellular sorting and trafficking of kAE1. We also demonstrated colocalization of kAE1 and basolateral-related sorting proteins in human kidney tissues by double immunofluorescence staining. These findings indicate that AP-1 mu1A, AP-3 mu1, AP-4 mu1 and clathrin are required for kAE1 sorting and trafficking from TGN to the basolateral membrane of acid-secreting α-intercalated cells.
Collapse
Affiliation(s)
- Mutita Junking
- Division of Molecular Medicine, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
20
|
Parmar HB, Barry C, Kai F, Duncan R. Golgi complex-plasma membrane trafficking directed by an autonomous, tribasic Golgi export signal. Mol Biol Cell 2014; 25:866-78. [PMID: 24451258 PMCID: PMC3952855 DOI: 10.1091/mbc.e13-07-0364] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The first example of a cytosolic, membrane-proximal, tribasic motif required for Golgi export to the plasma membrane is identified and characterized. This novel Golgi export signal can also mediate trafficking of a heterologous Golgi-resident protein, indicating that it functions as an autonomous Golgi export signal. Although numerous linear motifs that direct protein trafficking within cells have been identified, there are few examples of linear sorting signals mediating directed export of membrane proteins from the Golgi complex to the plasma membrane. The reovirus fusion-associated small transmembrane proteins are simple, single-pass transmembrane proteins that traffic through the endoplasmic reticulum–Golgi pathway to the plasma membrane, where they induce cell–cell membrane fusion. Here we show that a membrane-proximal, polybasic motif (PBM) in the cytosolic tail of p14 is essential for efficient export of p14 from the Golgi complex to the plasma membrane. Extensive mutagenic analysis reveals that the number, but not the identity or position, of basic residues present in the PBM dictates p14 export from the Golgi complex, with a minimum of three basic residues required for efficient Golgi export. Results further indicate that the tribasic motif does not affect plasma membrane retention of p14. Furthermore, introduction of the tribasic motif into a Golgi-localized, chimeric ERGIC-53 protein directs export from the Golgi complex to the plasma membrane. The p14 PBM is the first example of an autonomous, tribasic signal required for Golgi export to the plasma membrane.
Collapse
Affiliation(s)
- Hirendrasinh B Parmar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | | | | |
Collapse
|
21
|
Szalinski CM, Labilloy A, Bruns JR, Weisz OA. VAMP7 modulates ciliary biogenesis in kidney cells. PLoS One 2014; 9:e86425. [PMID: 24466086 PMCID: PMC3899255 DOI: 10.1371/journal.pone.0086425] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/12/2013] [Indexed: 12/02/2022] Open
Abstract
Epithelial cells elaborate specialized domains that have distinct protein and lipid compositions, including the apical and basolateral surfaces and primary cilia. Maintaining the identity of these domains is required for proper cell function, and requires the efficient and selective SNARE-mediated fusion of vesicles containing newly synthesized and recycling proteins with the proper target membrane. Multiple pathways exist to deliver newly synthesized proteins to the apical surface of kidney cells, and the post-Golgi SNAREs, or VAMPs, involved in these distinct pathways have not been identified. VAMP7 has been implicated in apical protein delivery in other cell types, and we hypothesized that this SNARE would have differential effects on the trafficking of apical proteins known to take distinct routes to the apical surface in kidney cells. VAMP7 expressed in polarized Madin Darby canine kidney cells colocalized primarily with LAMP2-positive compartments, and siRNA-mediated knockdown modulated lysosome size, consistent with the known function of VAMP7 in lysosomal delivery. Surprisingly, VAMP7 knockdown had no effect on apical delivery of numerous cargoes tested, but did decrease the length and frequency of primary cilia. Additionally, VAMP7 knockdown disrupted cystogenesis in cells grown in a three-dimensional basement membrane matrix. The effects of VAMP7 depletion on ciliogenesis and cystogenesis are not directly linked to the disruption of lysosomal function, as cilia lengths and cyst morphology were unaffected in an MDCK lysosomal storage disorder model. Together, our data suggest that VAMP7 plays an essential role in ciliogenesis and lumen formation. To our knowledge, this is the first study implicating an R-SNARE in ciliogenesis and cystogenesis.
Collapse
Affiliation(s)
- Christina M. Szalinski
- Renal Electrolyte Division, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - Anatália Labilloy
- Renal Electrolyte Division, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
- Ciência sem Fronteiras, CNPq, Brasilia, Brazil
| | - Jennifer R. Bruns
- Renal Electrolyte Division, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - Ora A. Weisz
- Renal Electrolyte Division, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Suzuki T, Orba Y, Makino Y, Okada Y, Sunden Y, Hasegawa H, Hall WW, Sawa H. Viroporin activity of the JC polyomavirus is regulated by interactions with the adaptor protein complex 3. Proc Natl Acad Sci U S A 2013; 110:18668-73. [PMID: 24167297 PMCID: PMC3832026 DOI: 10.1073/pnas.1311457110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Viroporins, which are encoded by a wide range of animal viruses, oligomerize in host cell membranes and form hydrophilic pores that can disrupt a number of physiological properties of the cell. Little is known about the relationship between host cell proteins and viroporin activity. The human JC polyomavirus (JCV) is the causative agent of progressive multifocal leukoencephalopathy. The JCV-encoded agnoprotein, which is essential for viral replication, has been shown to act as a viroporin. Here we demonstrate that the JCV agnoprotein specifically interacts with adaptor protein complex 3 through its δ subunit. This interaction interrupts adaptor protein complex 3-mediated vesicular trafficking with suppression of the targeting of the protein to the lysosomal degradation pathway and instead permits the transport of agnoprotein to the cell surface with resulting membrane permeabilization. The findings demonstrate a previously undescribed paradigm in virus-host interactions allowing the host to regulate viroporin activity and suggest that the viroporins of other viruses may also be highly regulated by specific interactions with host cell proteins.
Collapse
Affiliation(s)
- Tadaki Suzuki
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- Department of Pathology, National Institute of Infectious Diseases, Musashimurayama 208-0011, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Yoshinori Makino
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Yuki Okada
- Laboratory of Molecular Cellular Pathology, Hokkaido University School of Medicine, Sapporo 060-8638, Japan
| | - Yuji Sunden
- Laboratory of Comparative Pathology, Hokkaido University School of Veterinary Medicine, Sapporo 060-0818, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Musashimurayama 208-0011, Japan
- Global Virus Network, Baltimore, MD 21201
| | - William W. Hall
- Global Virus Network, Baltimore, MD 21201
- Centre for Research in Infectious Diseases, University College Dublin, Dublin 4, Ireland; and
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- Global Virus Network, Baltimore, MD 21201
- Global Centers of Excellence Program for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| |
Collapse
|
23
|
Hu G, Suo Y, Huang J. A crucial role of the RGS domain in trans-Golgi network export of AtRGS1 in the protein secretory pathway. MOLECULAR PLANT 2013; 6:1933-1944. [PMID: 23793400 DOI: 10.1093/mp/sst109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The secretory pathway is responsible for the transport of newly synthesized transmembrane proteins from the endoplasmic reticulum to their destinations via the Golgi/trans-Golgi network (TGN). Cargo proteins at each station are actively sorted by specific sorting signals on the cargo and the corresponding coat complexes. Here, we used the Arabidopsis regulator of G-protein signaling (AtRGS1), which contains an N-terminal potentially sensing glucose seven-transmembrane domain and a C-terminal RGS domain, as a model to uncover sorting motifs required for its cell surface expression. Expression of wild-type and truncated or mutated AtRGS1 fluorescent fusion proteins identified two cysteine residues in the extracellular N-terminus that are essential for endoplasmic reticulum exit and/or correct folding of AtRGS1. The linker between the seven-transmembrane and RGS domains contains an endoplasmic reticulum export signal, whereas the C-terminus is dispensable for the plasma membrane expression of AtRGS1. Interestingly, deletion of the RGS domain results in Golgi/TGN localization of the truncated AtRGS1. Further analysis using site-directed mutagenesis showed that a tyrosine-based motif embedded in the RGS domain is essential for Golgi/TGN export of AtRGS1. These results reveal a new role for the RGS domain in regulating AtRGS1 trafficking from the Golgi/TGN to the plasma membrane and explain the interaction between the seven-transmembrane and RGS domains.
Collapse
Affiliation(s)
- Guangzhen Hu
- National Key Laboratory of Plant Molecular Genetics, Shanghai Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 300 Feng Lin Road, Shanghai 200032, China
| | | | | |
Collapse
|
24
|
|
25
|
Abstract
Epithelial cells have an apical-basolateral axis of polarity, which is required for epithelial functions including barrier formation, vectorial ion transport and sensory perception. Here we review what is known about the sorting signals, machineries and pathways that maintain this asymmetry, and how polarity proteins interface with membrane-trafficking pathways to generate membrane domains de novo. It is becoming apparent that membrane traffic does not simply reinforce polarity, but is critical for the generation of cortical epithelial cell asymmetry.
Collapse
|
26
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
27
|
Carvajal-Gonzalez JM, Gravotta D, Mattera R, Diaz F, Perez Bay A, Roman AC, Schreiner RP, Thuenauer R, Bonifacino JS, Rodriguez-Boulan E. Basolateral sorting of the coxsackie and adenovirus receptor through interaction of a canonical YXXPhi motif with the clathrin adaptors AP-1A and AP-1B. Proc Natl Acad Sci U S A 2012; 109:3820-5. [PMID: 22343291 PMCID: PMC3309744 DOI: 10.1073/pnas.1117949109] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The coxsackie and adenovirus receptor (CAR) plays key roles in epithelial barrier function at the tight junction, a localization guided in part by a tyrosine-based basolateral sorting signal, (318)YNQV(321). Sorting motifs of this type are known to route surface receptors into clathrin-mediated endocytosis through interaction with the medium subunit (μ2) of the clathrin adaptor AP-2, but how they guide new and recycling membrane proteins basolaterally is unknown. Here, we show that YNQV functions as a canonical YxxΦ motif, with both Y318 and V321 required for the correct basolateral localization and biosynthetic sorting of CAR, and for interaction with a highly conserved pocket in the medium subunits (μ1A and μ1B) of the clathrin adaptors AP-1A and AP-1B. Knock-down experiments demonstrate that AP-1A plays a role in the biosynthetic sorting of CAR, complementary to the role of AP-1B in basolateral recycling of this receptor. Our study illustrates how two clathrin adaptors direct basolateral trafficking of a plasma membrane protein through interaction with a canonical YxxΦ motif.
Collapse
Affiliation(s)
- Jose Maria Carvajal-Gonzalez
- Department of Ophthalmology, Department of Cell and Developmental Biology, Department of Physiology and Biophysics, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065
| | - Diego Gravotta
- Department of Ophthalmology, Department of Cell and Developmental Biology, Department of Physiology and Biophysics, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065
| | - Rafael Mattera
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Fernando Diaz
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Andres Perez Bay
- Department of Ophthalmology, Department of Cell and Developmental Biology, Department of Physiology and Biophysics, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065
| | - Angel C. Roman
- Instituto Cajal–Consejo Superior de Investigaciones Cientificas, 28002 Madrid, Spain; and
| | - Ryan P. Schreiner
- Department of Ophthalmology, Department of Cell and Developmental Biology, Department of Physiology and Biophysics, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065
| | | | - Juan S. Bonifacino
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Enrique Rodriguez-Boulan
- Department of Ophthalmology, Department of Cell and Developmental Biology, Department of Physiology and Biophysics, Margaret Dyson Vision Research Institute, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
28
|
Anterograde trafficking of nascent α(2B)-adrenergic receptor: structural basis, roles of small GTPases. CURRENT TOPICS IN MEMBRANES 2012; 67:79-100. [PMID: 21771486 DOI: 10.1016/b978-0-12-384921-2.00004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
|
29
|
Abstract
Anterograde trafficking of newly synthesized G protein-coupled -receptors (GPCRs) from the endoplasmic reticulum to the cell surface represents a crucial checkpoint in controlling the amount of the functional receptors at the cell surface and the strength of signaling initiated by the receptors. In contrast to the extensively studied, well-understood endocytic and recycling pathways, the molecular mechanisms underlying the cell-surface targeting of the receptors remain poorly defined. In this chapter, I will discuss current advances in understanding post-Golgi transport of GPCRs by focusing on specific motifs or sequences that may function as sorting signals regulating export from the Golgi and subsequent transport to the plasma membrane of GPCRs.
Collapse
|
30
|
Wang G, Wu G. Small GTPase regulation of GPCR anterograde trafficking. Trends Pharmacol Sci 2011; 33:28-34. [PMID: 22015208 DOI: 10.1016/j.tips.2011.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 01/14/2023]
Abstract
The physiological functions of heterotrimeric G protein-coupled receptors (GPCRs) are dictated by their intracellular trafficking and precise targeting to the functional destinations. Over the past decades, most studies on the trafficking of GPCRs have focused on the events involved in endocytosis and recycling. By contrast, the molecular mechanisms underlying anterograde transport of newly synthesized GPCRs from the endoplasmic reticulum (ER) to the cell surface have only now begun to be revealed. In this review we discuss current advances in understanding the role of Ras-like GTPases, specifically the Rab and Sar1/ARF subfamilies, in regulating cell-surface transport of GPCRs en route from the ER and the Golgi.
Collapse
Affiliation(s)
- Guansong Wang
- Institute of Respiratory Diseases, Second Affiliated Hospital of the Third Military Medical University, Chongqing 400037, China
| | | |
Collapse
|
31
|
Zhang X, Dong C, Wu QJ, Balch WE, Wu G. Di-acidic motifs in the membrane-distal C termini modulate the transport of angiotensin II receptors from the endoplasmic reticulum to the cell surface. J Biol Chem 2011; 286:20525-35. [PMID: 21507945 PMCID: PMC3121450 DOI: 10.1074/jbc.m111.222034] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 04/08/2011] [Indexed: 12/18/2022] Open
Abstract
The molecular mechanisms underlying the endoplasmic reticulum (ER) export and cell surface transport of nascent G protein-coupled receptors (GPCRs) have just begun to be revealed and previous studies have shown that hydrophobic motifs in the putative amphipathic 8(th) α-helical region within the membrane-proximal C termini play an important role. In this study, we demonstrate that di-acidic motifs in the membrane-distal, nonstructural C-terminal portions are required for the exit from the ER and transport to the plasma membrane of angiotensin II receptors, but not adrenergic receptors. More interestingly, distinct di-acidic motifs dictate optimal export trafficking of different angiotensin II receptors and export ability of each acidic residue in the di-acidic motifs cannot be fully substituted by other acidic residue. Moreover, the function of the di-acidic motifs is likely mediated through facilitating the recruitment of the receptors onto the ER-derived COPII transport vesicles. Therefore, the di-acidic motifs located in the membrane-distal C termini may represent the first linear motifs which recruit selective GPCRs onto the COPII vesicles to control their export from the ER.
Collapse
Affiliation(s)
- Xiaoping Zhang
- From the Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Chunmin Dong
- From the Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Qiong J. Wu
- From the Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - William E. Balch
- the Department of Cell Biology, Scripps Research Institute, La Jolla, California 92037, and
| | - Guangyu Wu
- From the Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- the Department of Pharmacology and Toxicology, Georgia Health Sciences University, Augusta, Georgia 30912
| |
Collapse
|
32
|
Luo W, Wang Y, Reiser G. Proteinase-activated receptors, nucleotide P2Y receptors, and μ-opioid receptor-1B are under the control of the type I transmembrane proteins p23 and p24A in post-Golgi trafficking. J Neurochem 2011; 117:71-81. [DOI: 10.1111/j.1471-4159.2011.07173.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Opposing effects of a tyrosine-based sorting motif and a PDZ-binding motif regulate human T-lymphotropic virus type 1 envelope trafficking. J Virol 2010; 84:6995-7004. [PMID: 20463077 DOI: 10.1128/jvi.01853-09] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) envelope (Env) glycoprotein mediates binding of the virus to its receptor on the surface of target cells and subsequent fusion of virus and cell membranes. To better understand the mechanisms that control HTLV-1 Env trafficking and activity, we have examined two protein-protein interaction motifs in the cytoplasmic domain of Env. One is the sequence YSLI, which matches the consensus YXXPhi motifs that are known to interact with various adaptor protein complexes; the other is the sequence ESSL at the C terminus of Env, which matches the consensus PDZ-binding motif. We show here that mutations that destroy the YXXPhi motif increased Env expression on the cell surface and increased cell-cell fusion activity. In contrast, mutation of the PDZ-binding motif greatly diminished Env expression in cells, which could be restored to wild-type levels either by mutating the YXXPhi motif or by silencing AP2 and AP3, suggesting that interactions with PDZ proteins oppose an Env degradation pathway mediated by AP2 and AP3. Silencing of the PDZ protein hDlg1 did not affect Env expression, suggesting that hDlg1 is not a binding partner for Env. Substitution of the YSLI sequence in HTLV-1 Env with YXXPhi elements from other cell or virus membrane-spanning proteins resulted in alterations in Env accumulation in cells, incorporation into virions, and virion infectivity. Env variants containing YXXPhi motifs that are predicted to have high-affinity interaction with AP2 accumulated to lower steady-state levels. Interestingly, mutations that destroy the YXXPhi motif resulted in viruses that were not infectious by cell-free or cell-associated routes of infection. Unlike YXXPhi, the function of the PDZ-binding motif manifests itself only in the producer cells; AP2 silencing restored the incorporation of PDZ-deficient Env into virus-like particles (VLPs) and the infectivity of these VLPs to wild-type levels.
Collapse
|
34
|
Danglot L, Chaineau M, Dahan M, Gendron MC, Boggetto N, Perez F, Galli T. Role of TI-VAMP and CD82 in EGFR cell-surface dynamics and signaling. J Cell Sci 2010; 123:723-35. [PMID: 20144992 DOI: 10.1242/jcs.062497] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The v-SNARE TI-VAMP (VAMP7) mediates exocytosis during neuritogenesis, phagocytosis and lysosomal secretion. It localizes to endosomes and lysosomes but also to the trans-Golgi network. Here we show that depletion of TI-VAMP enhances the endocytosis of activated EGF receptor (EGFR) without affecting constitutive endocytosis of EGFR, or transferrin uptake. This increased EGFR internalization is mainly clathrin dependent. Searching for defects in EGFR regulators, we found that TI-VAMP depletion reduces the cell surface amount of CD82, a tetraspanin known to control EGFR localization in microdomains. We further show that TI-VAMP is required for secretion from the Golgi apparatus to the cell surface, and that TI-VAMP-positive vesicles transport CD82. Quantum dots video-microscopy indicates that depletion of TI-VAMP, or its cargo CD82, restrains EGFR diffusion and the area explored by EGFR at the cell surface. Both depletions also impair MAPK signaling and enhance endocytosis of activated EGFR by increased recruitment of AP-2. These results highlight the role of TI-VAMP in the secretory pathway of a tetraspanin, and support a model in which CD82 allows EGFR entry in microdomains that control its clathrin-dependent endocytosis and signaling.
Collapse
Affiliation(s)
- Lydia Danglot
- INSERM U950, Membrane Traffic in Neuronal & Epithelial Morphogenesis, Paris, F-75013, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Lang SM, Means RE. Characterization of cytoplasmic motifs important in rhesus rhadinovirus gB processing and trafficking. Virology 2010; 398:233-42. [PMID: 20060555 DOI: 10.1016/j.virol.2009.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 11/09/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022]
Abstract
Rhesus monkey rhadinovirus (RRV) is highly related to Kaposi's sarcoma-associated herpesvirus (KSHV), a human gamma-herpesvirus etiologically-linked with several cancers. Glycoprotein B (gB) homologues are encoded by all herpesviruses and play a role in virus attachment, entry, and in egress. We have found that RRV gB, like KSHV gB, is cleaved at a consensus furin cleavage site and is modified by both N-linked and O-linked glycosylation. Mutagenesis of three tyrosine- based trafficking motifs, a diacidic tyrosine motif, and a di-lucine motif in the cytoplasmic region revealed a role for these sequences in both ER export and endocytosis from the plasma membrane. These experiments provide a basis for further experiments looking at gB incorporation and role in gamma-herpesvirus assembly and egress.
Collapse
Affiliation(s)
- Sabine M Lang
- Department Of Pathology, Yale University School of Medicine, P.O. Box 208023, New Haven, CT 06520, USA
| | | |
Collapse
|
36
|
von Blume J, Duran JM, Forlanelli E, Alleaume AM, Egorov M, Polishchuk R, Molina H, Malhotra V. Actin remodeling by ADF/cofilin is required for cargo sorting at the trans-Golgi network. ACTA ACUST UNITED AC 2009; 187:1055-69. [PMID: 20026655 PMCID: PMC2806282 DOI: 10.1083/jcb.200908040] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sorting of both soluble and integral membrane proteins is disrupted by loss of ADF/cofilin, suggesting that actin severing controls expansion of a sorting domain within the TGN. Knockdown of the actin-severing protein actin-depolymerizing factor (ADF)/cofilin inhibited export of an exogenously expressed soluble secretory protein from Golgi membranes in Drosophilamelanogaster and mammalian tissue culture cells. A stable isotope labeling by amino acids in cell culture mass spectrometry–based protein profiling revealed that a large number of endogenous secretory proteins in mammalian cells were not secreted upon ADF/cofilin knockdown. Although many secretory proteins were retained, a Golgi-resident protein and a lysosomal hydrolase were aberrantly secreted upon ADF/cofilin knockdown. Overall, our findings indicate that inactivation of ADF/cofilin perturbed the sorting of a subset of both soluble and integral membrane proteins at the trans-Golgi network (TGN). We suggest that ADF/cofilin-dependent actin trimming generates a sorting domain at the TGN, which filters secretory cargo for export, and that uncontrolled growth of this domain causes missorting of proteins. This type of actin-dependent compartmentalization and filtering of secretory cargo at the TGN by ADF/cofilin could explain sorting of proteins that are destined to the cell surface.
Collapse
Affiliation(s)
- Julia von Blume
- Department of Cell and Developmental Biology, Centre de Regulació Genòmica, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Gonzalez A, Rodriguez-Boulan E. Clathrin and AP1B: key roles in basolateral trafficking through trans-endosomal routes. FEBS Lett 2009; 583:3784-95. [PMID: 19854182 DOI: 10.1016/j.febslet.2009.10.050] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/15/2009] [Accepted: 10/20/2009] [Indexed: 12/12/2022]
Abstract
Research following introduction of the MDCK model system to study epithelial polarity (1978) led to an initial paradigm that posited independent roles of the trans Golgi network (TGN) and recycling endosomes (RE) in the generation of, respectively, biosynthetic and recycling routes of plasma membrane (PM) proteins to apical and basolateral PM domains. This model dominated the field for 20 years. However, studies over the past decade and the discovery of the involvement of clathrin and clathrin adaptors in protein trafficking to the basolateral PM has led to a new paradigm. TGN and RE are now believed to cooperate closely in both biosynthetic and recycling trafficking routes. Here, we critically review these recent advances and the questions that remain unanswered.
Collapse
Affiliation(s)
- Alfonso Gonzalez
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Centro de Regulación Celular y Patología and Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile.
| | | |
Collapse
|
38
|
Fölsch H, Mattila PE, Weisz OA. Taking the scenic route: biosynthetic traffic to the plasma membrane in polarized epithelial cells. Traffic 2009; 10:972-81. [PMID: 19453969 PMCID: PMC2786770 DOI: 10.1111/j.1600-0854.2009.00927.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The maintenance of epithelial cell function requires the establishment and continuous renewal of differentiated apical and basolateral plasma membrane domains with distinct lipid and protein compositions. Newly synthesized proteins destined for either surface domain are processed along the biosynthetic pathway and segregated into distinct subsets of transport carriers emanating from the trans-Golgi network. Recent studies have illuminated additional complexities in the subsequent delivery of these proteins to the cell surface. In particular, multiple routes to the apical and basolateral cell surfaces have been uncovered, and many of these involve indirect passage through endocytic compartments. This review summarizes our current understanding of these routes and discusses open issues that remain to be clarified.
Collapse
Affiliation(s)
- Heike Fölsch
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, 2205 Tech Drive, Evanston, IL 60208
| | - Polly E. Mattila
- Department of Medicine University of Pittsburgh, 978.1 Scaife Hall, 3550 Terrace St., Pittsburgh, PA 15261
| | - Ora A. Weisz
- Department of Medicine University of Pittsburgh, 978.1 Scaife Hall, 3550 Terrace St., Pittsburgh, PA 15261
- Cell Biology and Physiology, University of Pittsburgh 978.1 Scaife Hall, 3550 Terrace St., Pittsburgh, PA 15261
| |
Collapse
|
39
|
Liersch T, Grade M, Gaedcke J, Varma S, Difilippantonio MJ, Langer C, Hess CF, Becker H, Ried T, Ghadimi BM. Preoperative chemoradiotherapy in locally advanced rectal cancer: correlation of a gene expression-based response signature with recurrence. CANCER GENETICS AND CYTOGENETICS 2009; 190:57-65. [PMID: 19380020 PMCID: PMC2766806 DOI: 10.1016/j.cancergencyto.2008.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 11/10/2008] [Accepted: 11/10/2008] [Indexed: 10/20/2022]
Abstract
Preoperative chemoradiotherapy is recommended for locally advanced rectal cancer (UICC stage II/III). We recently demonstrated that responsive and nonresponsive tumors showed differential expression levels of 54 genes. In this follow-up study, we investigated the relationship between this gene set and disease-free (DFS) and overall survival (OS). Pretherapeutic biopsies from 30 participants in the CAO/ARO/AIO-94 trial of the German Rectal Cancer Study Group were analyzed using gene expression microarrays. Statistical analysis was performed to identify differentially expressed genes between recurrent and nonrecurrent tumors and to correlate these changes with disease recurrence and outcome. After a median follow-up of 59 months, seven of eight patients with recurrent disease was a nonresponder, and one responsive tumor recurred. Response to chemoradiotherapy was significantly correlated with an improved DFS (log rank P=0.028), whereas OS did not differ significantly (P=0.11). Applying a class comparison analysis, we identified 20 genes that were differentially expressed between recurrent and nonrecurrent tumors (P<0.001). Analyzing the first two principal components of the 54 genes previously identified to predict response, we observed that this response signature correlated with an increased risk of cancer recurrence. These data suggest that the genetic basis of local response also affects the genetic basis of tumor recurrence. Genes that are indicative of nonresponse to preoperative chemoradiotherapy might also be linked to an increased risk of tumor recurrence.
Collapse
Affiliation(s)
- Torsten Liersch
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
| | - Marian Grade
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Jochen Gaedcke
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
| | - Sudhir Varma
- Biometrics Research Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | | | - Claus Langer
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
| | - Clemens F. Hess
- Department of Radiation Oncology and Radiotherapy, University Medical Center, Göttingen, Germany
| | - Heinz Becker
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
| | - Thomas Ried
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A
| | - B. Michael Ghadimi
- Department of General and Visceral Surgery, University Medical Center, Göttingen, Germany
| |
Collapse
|
40
|
Reovirus FAST protein transmembrane domains function in a modular, primary sequence-independent manner to mediate cell-cell membrane fusion. J Virol 2009; 83:2941-50. [PMID: 19129451 DOI: 10.1128/jvi.01869-08] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The FAST proteins are a unique family of virus-encoded cell-cell membrane fusion proteins. In the absence of a cleavable N-terminal signal peptide, a single-pass transmembrane domain (TMD) functions as a reverse signal-anchor to direct the FAST proteins into the plasma membrane in an N(exo)/C(cyt) topology. There is little information available on the role of the FAST protein TMD in the cell-cell membrane fusion reaction. We show that in the absence of conservation in the length or primary amino acid sequence, the p14 TMD can be functionally exchanged with the TMDs of the p10 and p15 FAST proteins. This is not the case for chimeric p14 proteins containing the TMDs of two different enveloped viral fusion proteins or a cellular membrane protein; such chimeric proteins were defective for both pore formation and syncytiogenesis. TMD structural features that are conserved within members of the FAST protein family presumably play direct roles in the fusion reaction. Molecular modeling suggests that the funnel-shaped architecture of the FAST protein TMDs may represent such a conserved structural and functional motif. Interestingly, although heterologous TMDs exert diverse influences on the trafficking of the p14 FAST protein, these TMDs are capable of functioning as reverse signal-anchor sequences to direct p14 into lipid rafts in the correct membrane topology. The FAST protein TMDs are therefore not primary determinants of type III protein topology, but they do play a direct, sequence-independent role in the membrane fusion reaction.
Collapse
|
41
|
Human immunodeficiency virus type-1 gag and host vesicular trafficking pathways. Curr Top Microbiol Immunol 2009; 339:67-84. [PMID: 20012524 DOI: 10.1007/978-3-642-02175-6_4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The Gag protein of HIV-1 directs the particle assembly process. Gag recruits components of the cellular vesicular trafficking machinery in order to traverse the cytoplasm of the cell and reach the particle assembly site. The plasma membrane is the primary site of particle assembly in most cell types, while in macrophages an unusual intracellular membrane-bound compartment bearing markers of late endosomes and the plasma membrane is the predominant assembly site. Plasma membrane specificity of assembly may be directed by components of lipid rafts and the cytoplasmic leaflet component PI(4,5)P(2). Recent work has highlighted the role of adaptor protein complexes, protein sorting and recycling pathways, components of the multivesicular body, and cellular motor proteins in facilitating HIV assembly and budding. This review presents an overview of the relevant vesicular trafficking pathways and describes the individual components implicated in interactions with Gag.
Collapse
|
42
|
Chapuy B, Tikkanen R, Mühlhausen C, Wenzel D, von Figura K, Höning S. AP-1 and AP-3 Mediate Sorting of Melanosomal and Lysosomal Membrane Proteins into Distinct Post-Golgi Trafficking Pathways. Traffic 2008; 9:1157-72. [DOI: 10.1111/j.1600-0854.2008.00745.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
43
|
A transplanted NPVY sequence in the cytosolic domain of the erythropoietin receptor enhances maturation. Biochem J 2008; 410:409-16. [PMID: 17995455 DOI: 10.1042/bj20071297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Activation of the EPO-R [EPO (erythropoietin) receptor] by its ligand EPO promotes erythropoiesis. Low cell surface EPO-R levels are traditionally attributed to inefficient folding mediated by the receptor extracellular domain. In the present study, we addressed the role of the EPO-R intracellular domain in exit from the ER (endoplasmic reticulum) and surface expression. A fusion protein between the thermo-reversible folding mutant of VSVG (vesicular-stomatitis-virus glycoprotein) (VSVGtsO45) and the EPO-R cytosolic domain [VSVG-WT (wild-type)] displayed delayed intracellular trafficking as compared with the parental VSVGtsO45, suggesting that the EPO-R cytosolic domain can hamper ER exit. Although NPXY-based motifs were originally associated with clathrin binding and endocytosis, they may also function in other contexts of the secretory pathway. A fusion protein between VSVGtsO45 and the cytosolic portion of EPO-R containing an NPVY insert (VSVG-NPVY) displayed enhanced glycan maturation and surface expression as compared with VSVG-WT. Notably, the NPVY insert also conferred improved maturation and augmented cell surface EPO-R. Our findings highlight three major concepts: (i) the EPO-R cytosolic domain is involved in ER exit of the receptor. (ii) Sequence motifs that participate in endocytosis can also modulate transport along the secretory pathway. (iii) VSVG-fusion proteins may be employed to screen for intracellular sequences that regulate transport.
Collapse
|
44
|
Nishimura N, Sasaki T. Cell-surface biotinylation to study endocytosis and recycling of occludin. Methods Mol Biol 2008; 440:89-96. [PMID: 18369939 DOI: 10.1007/978-1-59745-178-9_7] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The dynamic turnover of adherens junctions (AJs) and tight junctions (TJs) is essential for epithelial morphogenesis during normal development and differentiation. Although the endocytic recycling of E-cadherin is characterized and implicated in AJ turnover, the molecular basis for TJ turnover is poorly understood. Occludin and claudins are distinct transmembrane proteins localized to the TJs. Although claudins are an indispensable structural component of TJ strands, depletion of occludin in mice reveals well-developed TJ strands and complex histological abnormalities. To examine the intracellular transport of transmembrane proteins to and from the cell surface, cell-surface biotinylation is a proven powerful method. Using this method, we successfully demonstrated that occludin was endocytosed and recycled back to the cell surface in both fibroblastic baby hamster kidney (BHK) and epithelial MTD-1A cells. The endocytic recycling of occludin as well as the formation of functional TJs was dependent on Rab13 and a junctional Rab13-binding protein (JRAB)/molecule interacting with CasL-like 2 (MICAL-L2). We describe the method to study the intracellular transport of occludin to and from the cell surface in both fibroblastic and epithelial cells.
Collapse
Affiliation(s)
- Noriyuki Nishimura
- Department of Biochemistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | | |
Collapse
|
45
|
Cancino J, Torrealba C, Soza A, Yuseff MI, Gravotta D, Henklein P, Rodriguez-Boulan E, González A. Antibody to AP1B adaptor blocks biosynthetic and recycling routes of basolateral proteins at recycling endosomes. Mol Biol Cell 2007; 18:4872-84. [PMID: 17881725 PMCID: PMC2096610 DOI: 10.1091/mbc.e07-06-0563] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 09/11/2007] [Indexed: 01/03/2023] Open
Abstract
The epithelial-specific adaptor AP1B sorts basolateral plasma membrane (PM) proteins in both biosynthetic and recycling routes, but the site where it carries out this function remains incompletely defined. Here, we have investigated this topic in Fischer rat thyroid (FRT) epithelial cells using an antibody against the medium subunit micro1B. This antibody was suitable for immunofluorescence and blocked the function of AP1B in these cells. The antibody blocked the basolateral recycling of two basolateral PM markers, Transferrin receptor (TfR) and LDL receptor (LDLR), in a perinuclear compartment with marker and functional characteristics of recycling endosomes (RE). Live imaging experiments demonstrated that in the presence of the antibody two newly synthesized GFP-tagged basolateral proteins (vesicular stomatitis virus G [VSVG] protein and TfR) exited the trans-Golgi network (TGN) normally but became blocked at the RE within 3-5 min. By contrast, the antibody did not block trafficking of green fluorescent protein (GFP)-LDLR from the TGN to the PM but stopped its recycling after internalization into RE in approximately 45 min. Our experiments conclusively demonstrate that 1) AP1B functions exclusively at RE; 2) TGN-to-RE transport is very fast and selective and is mediated by adaptors different from AP1B; and 3) the TGN and AP1B-containing RE cooperate in biosynthetic basolateral sorting.
Collapse
Affiliation(s)
- Jorge Cancino
- *Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Carolina Torrealba
- *Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Andrea Soza
- *Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - María Isabel Yuseff
- *Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Diego Gravotta
- Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10021
| | - Peter Henklein
- Institute of Biochemistry Faculty of Medicine, Humboldt University, 10117 Berlin, Germany; and
| | - Enrique Rodriguez-Boulan
- Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10021
| | - Alfonso González
- *Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| |
Collapse
|
46
|
Fields IC, Shteyn E, Pypaert M, Proux-Gillardeaux V, Kang RS, Galli T, Fölsch H. v-SNARE cellubrevin is required for basolateral sorting of AP-1B-dependent cargo in polarized epithelial cells. ACTA ACUST UNITED AC 2007; 177:477-88. [PMID: 17485489 PMCID: PMC2034334 DOI: 10.1083/jcb.200610047] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The epithelial cell–specific adaptor complex AP-1B is crucial for correct delivery of many transmembrane proteins from recycling endosomes to the basolateral plasma membrane. Subsequently, membrane fusion is dependent on the formation of complexes between SNARE proteins located at the target membrane and on transport vesicles. Although the t-SNARE syntaxin 4 has been localized to the basolateral membrane, the v-SNARE operative in the AP-1B pathway remained unknown. We show that the ubiquitously expressed v-SNARE cellubrevin localizes to the basolateral membrane and to recycling endosomes, where it colocalizes with AP-1B. Furthermore, we demonstrate that cellubrevin coimmunoprecipitates preferentially with syntaxin 4, implicating this v-SNARE in basolateral fusion events. Cleavage of cellubrevin with tetanus neurotoxin (TeNT) results in scattering of AP-1B localization and missorting of AP-1B–dependent cargos, such as transferrin receptor and a truncated low-density lipoprotein receptor, LDLR-CT27. These data suggest that cellubrevin and AP-1B cooperate in basolateral membrane trafficking.
Collapse
Affiliation(s)
- Ian C Fields
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Newell-Litwa K, Seong E, Burmeister M, Faundez V. Neuronal and non-neuronal functions of the AP-3 sorting machinery. J Cell Sci 2007; 120:531-41. [PMID: 17287392 DOI: 10.1242/jcs.03365] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vesicles selectively exchange lipids, membrane proteins and luminal contents between organelles along the exocytic and endocytic routes. The repertoire of membrane proteins present in these vesicles is crucial for their targeting and function. Vesicle composition is determined at the time of their biogenesis by cytosolic coats. The heterotetrameric protein adaptor protein complex 3 (AP-3), a coat component, participates in the generation of a diverse group of secretory organelles and lysosome-related organelles. Recent work has shed light on the mechanisms that regulate AP-3 and the trafficking pathways controlled by this adaptor. Phenotypic analysis of organisms carrying genetic deficiencies in the AP-3 pathway highlight its role regulating the targeting of lysosomal, melanosomal and synaptic vesicle-specific membrane proteins. Synaptic vesicles from AP-3-deficient mice possess altered levels of neurotransmitter and ion transporters, molecules that ultimately define the type and amount of neurotransmitter stored in these vesicles. These findings reveal a complex picture of how AP-3 functions in multiple tissues, including neuronal tissue, and expose potential links between endocytic sorting mechanisms and the pathogenesis of psychiatric disorders such as schizophrenia.
Collapse
|
48
|
Gravotta D, Deora A, Perret E, Oyanadel C, Soza A, Schreiner R, Gonzalez A, Rodriguez-Boulan E. AP1B sorts basolateral proteins in recycling and biosynthetic routes of MDCK cells. Proc Natl Acad Sci U S A 2007; 104:1564-9. [PMID: 17244703 PMCID: PMC1785260 DOI: 10.1073/pnas.0610700104] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The epithelial-specific adaptor AP1B sorts basolateral proteins, but the trafficking routes where it performs its sorting role remain controversial. Here, we used an RNAi approach to knock down the medium subunit of AP1B (mu1B) in the prototype epithelial cell line Madin-Darby canine kidney (MDCK). Mu1B-knocked down MDCK cells displayed loss of polarity of several endogenous and exogenous basolateral markers transduced via adenovirus vectors, but exhibited normal polarity of apical markers. We chose two well characterized basolateral protein markers, the transferrin receptor (TfR) and the vesicular stomatitis virus G protein, to study the sorting role of AP1B. A surface-capture assay introduced here showed that mu1B-knocked down MDCK cells plated on filters at confluency and cultured for 4.5 d, sorted TfR correctly in the biosynthetic route but incorrectly in the recycling route. In contrast, these same cells missorted vesicular stomatitis virus G apically in the biosynthetic route. Strikingly, recently confluent MDCK cells (1-3 d) displayed AP1B-dependence in the biosynthetic route of TfR, which decreased with additional days in culture. Sucrose density gradient analysis detected AP1B predominantly in TfR-rich endosomal fractions in MDCK cells confluent for 1 and 4 d. Our results are consistent with the following model: AP1B sorts basolateral proteins in both biosynthetic and recycling routes of MDCK cells, as a result of its predominant functional localization in recycling endosomes, which constitute a post-Golgi station in the biosynthetic route of some plasma membrane proteins. TfR utilizes a direct route from Golgi to basolateral membrane that is established as the epithelial monolayer matures.
Collapse
Affiliation(s)
- Diego Gravotta
- *Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021
| | - Ami Deora
- *Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021
| | - Emilie Perret
- *Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021
| | - Claudia Oyanadel
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile; and
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Andrea Soza
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile; and
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Ryan Schreiner
- *Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021
| | - Alfonso Gonzalez
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, and Centro de Regulación Celular y Patología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 6510260 Santiago, Chile; and
- Millennium Institute for Fundamental and Applied Biology, 7780344 Santiago, Chile
| | - Enrique Rodriguez-Boulan
- *Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
49
|
Dong C, Wu G. Regulation of anterograde transport of alpha2-adrenergic receptors by the N termini at multiple intracellular compartments. J Biol Chem 2006; 281:38543-54. [PMID: 17038316 PMCID: PMC2648813 DOI: 10.1074/jbc.m605734200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The studies on the intrinsic structural determinants for export trafficking of G protein-coupled receptors (GPCRs) have been mainly focused on the C termini of the receptors. In this report we determined the role of the extracellular N termini of alpha(2)-adrenergic receptors (alpha(2)-ARs) in the anterograde transport from the endoplasmic reticulum (ER) through the Golgi to the cell surface. The N-terminal-truncated alpha(2B)-AR mutant is completely unable to target to the cell surface. A single Met-6 residue is essential for the export of alpha(2B)-AR from the ER, likely through modulating correct alpha(2B)-AR folding in the ER. The Tyr-Ser motif, highly conserved in the membrane-proximal N termini of all alpha(2)-AR subtypes, is required for the exit of alpha(2A)-AR and alpha(2B)-AR from the Golgi apparatus, thus representing a novel Tyr-based motif modulating GPCR transport at the Golgi level. These data provide the first evidence indicating an essential role of the N termini of GPCRs in the export from distinct intracellular compartments along the secretory pathway.
Collapse
Affiliation(s)
- Chunmin Dong
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA
| | | |
Collapse
|
50
|
Dong C, Filipeanu CM, Duvernay MT, Wu G. Regulation of G protein-coupled receptor export trafficking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1768:853-70. [PMID: 17074298 PMCID: PMC1885203 DOI: 10.1016/j.bbamem.2006.09.008] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 09/14/2006] [Accepted: 09/18/2006] [Indexed: 12/26/2022]
Abstract
G protein-coupled receptors (GPCRs) constitute a superfamily of cell-surface receptors which share a common topology of seven transmembrane domains and modulate a variety of cell functions through coupling to heterotrimeric G proteins by responding to a vast array of stimuli. The magnitude of cellular response elicited by a given signal is dictated by the level of GPCR expression at the plasma membrane, which is the balance of elaborately regulated endocytic and exocytic trafficking. This review will cover recent advances in understanding the molecular mechanism underlying anterograde transport of the newly synthesized GPCRs from the endoplasmic reticulum (ER) through the Golgi to the plasma membrane. We will focus on recently identified motifs involved in GPCR exit from the ER and the Golgi, GPCR folding in the ER and the rescue of misfolded receptors from within, GPCR-interacting proteins that modulate receptor cell-surface targeting, pathways that mediate GPCR traffic, and the functional role of export in controlling GPCR signaling.
Collapse
Affiliation(s)
| | | | | | - Guangyu Wu
- * Corresponding author. Tel: +1 504 568 2236; Fax: +1 504 568 2361. E-mail address: (G. Wu)
| |
Collapse
|