1
|
Wang J, Fourriere L, Gleeson PA. Advances in the cell biology of the trafficking and processing of amyloid precursor protein: impact of familial Alzheimer's disease mutations. Biochem J 2024; 481:1297-1325. [PMID: 39302110 PMCID: PMC11555708 DOI: 10.1042/bcj20240056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
The production of neurotoxic amyloid-β peptides (Aβ) is central to the initiation and progression of Alzheimer's disease (AD) and involves sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. APP and the secretases are transmembrane proteins and their co-localisation in the same membrane-bound sub-compartment is necessary for APP cleavage. The intracellular trafficking of APP and the β-secretase, BACE1, is critical in regulating APP processing and Aβ production and has been studied in several cellular systems. Here, we summarise the intracellular distribution and transport of APP and its secretases, and the intracellular location for APP cleavage in non-polarised cells and neuronal models. In addition, we review recent advances on the potential impact of familial AD mutations on APP trafficking and processing. This is critical information in understanding the molecular mechanisms of AD progression and in supporting the development of novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Jingqi Wang
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Lou Fourriere
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A. Gleeson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
2
|
Healy MD, Collins BM. The PDLIM family of actin-associated proteins and their emerging role in membrane trafficking. Biochem Soc Trans 2023; 51:2005-2016. [PMID: 38095060 PMCID: PMC10754285 DOI: 10.1042/bst20220804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
The PDZ and LIM domain (PDLIM) proteins are associated with the actin cytoskeleton and have conserved in roles in metazoan actin organisation and function. They primarily function as scaffolds linking various proteins to actin and its binding partner α-actinin via two conserved domains; an N-terminal postsynaptic density 95, discs large and zonula occludens-1 (PDZ) domain, and either single or multiple C-terminal LIN-11, Isl-1 and MEC-3 (LIM) domains in the actinin-associated LIM protein (ALP)- and Enigma-related proteins, respectively. While their role in actin organisation, such as in stress fibres or in the Z-disc of muscle fibres is well known, emerging evidence also suggests a role in actin-dependent membrane trafficking in the endosomal system. This is mediated by a recently identified interaction with the sorting nexin 17 (SNX17) protein, an adaptor for the trafficking complex Commander which is itself intimately linked to actin-directed formation of endosomal recycling domains. In this review we focus on the currently understood structural basis for PDLIM function. The PDZ domains mediate direct binding to distinct classes of PDZ-binding motifs (PDZbms), including α-actinin and other actin-associated proteins, and a highly specific interaction with the type III PDZbm such as the one found in the C-terminus of SNX17. The structures of the LIM domains are less well characterised and how they engage with their ligands is completely unknown. Despite the lack of experimental structural data, we find that recently developed machine learning-based structure prediction methods provide insights into their potential interactions and provide a template for further studies of their molecular functions.
Collapse
Affiliation(s)
- Michael D. Healy
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| | - Brett M. Collins
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| |
Collapse
|
3
|
Sumya FT, Pokrovskaya ID, Lupashin VV. Rapid COG Depletion in Mammalian Cell by Auxin-Inducible Degradation System. Methods Mol Biol 2023; 2557:365-390. [PMID: 36512227 PMCID: PMC10019459 DOI: 10.1007/978-1-0716-2639-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Conserved oligomeric Golgi (COG) complex orchestrates intra-Golgi retrograde trafficking and glycosylation of macromolecules, but the detailed mechanism of COG action is unknown. Previous studies employed prolonged protein knockout and knockdown approaches which may potentially generate off-target and indirect mutant phenotypes. To achieve a fast depletion of COG subunits in human cells, the auxin-inducible degradation system was employed. This method of protein regulation allows a very fast and efficient depletion of COG subunits, which provides the ability to accumulate COG complex dependent (CCD) vesicles and investigate initial cellular defects associated with the acute depletion of COG complex subunits. This protocol is applicable to other vesicle tethering complexes and can be utilized to investigate anterograde and retrograde intracellular membrane trafficking pathways.
Collapse
Affiliation(s)
- Farhana Taher Sumya
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Irina D Pokrovskaya
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Vladimir V Lupashin
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
4
|
Houghton FJ, Makhoul C, Cho EHJ, Williamson NA, Gleeson PA. Interacting partners of Golgi-localized small G protein Arl5b identified by a combination of in vivo proximity labelling and GFP-Trap pull down. FEBS Lett 2022; 596:2382-2399. [PMID: 35789482 DOI: 10.1002/1873-3468.14443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/10/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
The small G protein Arl5b is localized on the trans-Golgi network (TGN) and regulates endosomes-to-TGN transport. Here, we combined in vivo and in vitro techniques to map the interactive partners and near neighbours of Arl5b at the TGN, using constitutively-active, membrane-bound Arl5b(Q70L)-GFP in stably expressing HeLa cells, and the proximity labelling techniques BioID and APEX2 in parallel with GFP-Trap pull-down. From mass spectrometry analysis, 22 Golgi proteins were identified; 50% were TGN-localised Rabs, Arfs and Arls. The scaffold/tethering factors ACBD3 (GCP60) and PIST (GOPC) were also identified, and we show that Arl5b is required for TGN recruitment of ACBD3. Overall, the combination of in vivo labelling and direct pull-downs indicates a highly organised complex of small G proteins on TGN membranes.
Collapse
Affiliation(s)
- Fiona J Houghton
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Christian Makhoul
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Ellie Hyun-Jung Cho
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.,Biological Optical Microscopy Platform, The University of Melbourne, Victoria, 3010, Australia
| | - Nicholas A Williamson
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
5
|
Carrà G, Avalle L, Seclì L, Brancaccio M, Morotti A. Shedding Light on NF-κB Functions in Cellular Organelles. Front Cell Dev Biol 2022; 10:841646. [PMID: 35620053 PMCID: PMC9127296 DOI: 10.3389/fcell.2022.841646] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
NF-κB is diffusely recognized as a transcriptional factor able to modulate the expression of various genes involved in a broad spectrum of cellular functions, including proliferation, survival and migration. NF-κB is, however, also acting outside the nucleus and beyond its ability to binds to DNA. NF-κB is indeed found to localize inside different cellular organelles, such as mitochondria, endoplasmic reticulum, Golgi and nucleoli, where it acts through different partners in mediating various biological functions. Here, we discuss the relationship linking NF-κB to the cellular organelles, and how this crosstalk between cellular organelles and NF-κB signalling may be evaluated for anticancer therapies.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| |
Collapse
|
6
|
An Updated View of the Importance of Vesicular Trafficking and Transport and Their Role in Immune-Mediated Diseases: Potential Therapeutic Interventions. MEMBRANES 2022; 12:membranes12060552. [PMID: 35736259 PMCID: PMC9230090 DOI: 10.3390/membranes12060552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022]
Abstract
Cellular trafficking is the set of processes of distributing different macromolecules by the cell. This process is highly regulated in cells, involving a system of organelles (endomembranous system), among which are a great variety of vesicles that can be secreted from the cell, giving rise to different types of extracellular vesicles (EVs) that can be captured by other cells to modulate their function. The cells of the immune system are especially sensitive to this cellular traffic, producing and releasing different classes of EVs, especially in disease states. There is growing interest in this field due to the therapeutic and translational possibilities it offers. Different ways of taking advantage of the understanding of cell trafficking and EVs are being investigated, and their use as biomarkers or therapeutic targets is being investigated. The objective of this review is to collect the latest results and knowledge in this area with a specific focus on immune-mediated diseases. Although some promising results have been obtained, further knowledge is still needed, at both the basic and translational levels, to understand and modulate cellular traffic and EVs for better clinical management of these patients.
Collapse
|
7
|
Sumya FT, Pokrovskaya ID, Lupashin V. Development and Initial Characterization of Cellular Models for COG Complex-Related CDG-II Diseases. Front Genet 2021; 12:733048. [PMID: 34603392 PMCID: PMC8484713 DOI: 10.3389/fgene.2021.733048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Conserved Oligomeric Golgi (COG) is an octameric protein complex that orchestrates intra-Golgi trafficking of glycosylation enzymes. Over a hundred individuals with 31 different COG mutations have been identified until now. The cellular phenotypes and clinical presentations of COG-CDGs are heterogeneous, and patients primarily represent neurological, skeletal, and hepatic abnormalities. The establishment of a cellular COG disease model will benefit the molecular study of the disease, explaining the detailed sequence of the interplay between the COG complex and the trafficking machinery. Moreover, patient fibroblasts are not a good representative of all the organ systems and cell types that are affected by COG mutations. We developed and characterized cellular models for human COG4 mutations, specifically in RPE1 and HEK293T cell lines. Using a combination of CRISPR/Cas9 and lentiviral transduction technologies, both myc-tagged wild-type and mutant (G516R and R729W) COG4 proteins were expressed under the endogenous COG4 promoter. Constructed isogenic cell lines were comprehensively characterized using biochemical, microscopy (superresolution and electron), and proteomics approaches. The analysis revealed similar stability and localization of COG complex subunits, wild-type cell growth, and normal Golgi morphology in all three cell lines. Importantly, COG4-G516R cells demonstrated increased HPA-647 binding to the plasma membrane glycoconjugates, while COG4-R729W cells revealed high GNL-647 binding, indicating specific defects in O- and N-glycosylation. Both mutant cell lines express an elevated level of heparin sulfate proteoglycans. Moreover, a quantitative mass-spectrometry analysis of proteins secreted by COG-deficient cell lines revealed abnormal secretion of SIL1 and ERGIC-53 proteins by COG4-G516R cells. Interestingly, the clinical phenotype of patients with congenital mutations in the SIL1 gene (Marinesco-Sjogren syndrome) overlaps with the phenotype of COG4-G516R patients (Saul-Wilson syndrome). Our work is the first compressive study involving the creation of different COG mutations in different cell lines other than the patient's fibroblast. It may help to address the underlying cause of the phenotypic defects leading to the discovery of a proper treatment guideline for COG-CDGs.
Collapse
Affiliation(s)
| | | | - Vladimir Lupashin
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
8
|
Navikas V, Leitao SM, Grussmayer KS, Descloux A, Drake B, Yserentant K, Werther P, Herten DP, Wombacher R, Radenovic A, Fantner GE. Correlative 3D microscopy of single cells using super-resolution and scanning ion-conductance microscopy. Nat Commun 2021; 12:4565. [PMID: 34315910 PMCID: PMC8316521 DOI: 10.1038/s41467-021-24901-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
High-resolution live-cell imaging is necessary to study complex biological phenomena. Modern fluorescence microscopy methods are increasingly combined with complementary, label-free techniques to put the fluorescence information into the cellular context. The most common high-resolution imaging approaches used in combination with fluorescence imaging are electron microscopy and atomic-force microscopy (AFM), originally developed for solid-state material characterization. AFM routinely resolves atomic steps, however on soft biological samples, the forces between the tip and the sample deform the fragile membrane, thereby distorting the otherwise high axial resolution of the technique. Here we present scanning ion-conductance microscopy (SICM) as an alternative approach for topographical imaging of soft biological samples, preserving high axial resolution on cells. SICM is complemented with live-cell compatible super-resolution optical fluctuation imaging (SOFI). To demonstrate the capabilities of our method we show correlative 3D cellular maps with SOFI implementation in both 2D and 3D with self-blinking dyes for two-color high-order SOFI imaging. Finally, we employ correlative SICM/SOFI microscopy for visualizing actin dynamics in live COS-7 cells with subdiffraction-resolution.
Collapse
Affiliation(s)
- Vytautas Navikas
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, Swiss Federal InstSIitute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Samuel M Leitao
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Kristin S Grussmayer
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, Swiss Federal InstSIitute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Grussmayer Lab, Department of Bionanoscience, Faculty of Applied Science and Kavli Institute for Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| | - Adrien Descloux
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, Swiss Federal InstSIitute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Barney Drake
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Klaus Yserentant
- College of Medical and Dental Sciences, Medical School & School of Chemistry, University of Birmingham, Birmingham, United Kingdom
| | - Philipp Werther
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Dirk-Peter Herten
- College of Medical and Dental Sciences, Medical School & School of Chemistry, University of Birmingham, Birmingham, United Kingdom
| | - Richard Wombacher
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Aleksandra Radenovic
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, Swiss Federal InstSIitute of Technology Lausanne (EPFL), Lausanne, Switzerland.
| | - Georg E Fantner
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
9
|
Sager G, Szul T, Lee E, Kawai R, Presley JF, Sztul E. Modeling the dynamic behaviors of the COPI vesicle formation regulators, the small GTPase Arf1 and its activating Sec7 guanine nucleotide exchange factor GBF1 on Golgi membranes. Mol Biol Cell 2021; 32:446-459. [PMID: 33405949 PMCID: PMC8098855 DOI: 10.1091/mbc.e20-09-0587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The components and subprocesses underlying the formation of COPI-coated vesicles at the Golgi are well understood. The coating cascade is initiated after the small GTPase Arf1 is activated by the Sec7 domain–containing guanine nucleotide exchange factor GBF1 (Golgi brefeldin A resistant guanine nucleotide exchange factor 1). This causes a conformational shift within Arf1 that facilitates stable association of Arf1 with the membrane, a process required for subsequent recruitment of the COPI coat. Although we have atomic-level knowledge of Arf1 activation by Sec7 domain–containing GEFs, our understanding of the biophysical processes regulating Arf1 and GBF1 dynamics is limited. We used fluorescence recovery after photobleaching data and kinetic Monte Carlo simulation to assess the behavior of Arf1 and GBF1 during COPI vesicle formation in live cells. Our analyses suggest that Arf1 and GBF1 associate with Golgi membranes independently, with an excess of GBF1 relative to Arf1. Furthermore, the GBF1-mediated Arf1 activation is much faster than GBF1 cycling on/off the membrane, suggesting that GBF1 is regulated by processes other than its interactions Arf1. Interestingly, modeling the behavior of the catalytically inactive GBF1/E794K mutant stabilized on the membrane is inconsistent with the formation of a stable complex between it and an endogenous Arf1 and suggests that GBF1/E794K is stabilized on the membrane independently of complex formation.
Collapse
Affiliation(s)
- Garrett Sager
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35924.,Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35924
| | - Tomasz Szul
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35924
| | - Eunjoo Lee
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35924
| | - Ryoichi Kawai
- Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35924
| | - John F Presley
- Department of Anatomy & Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Elizabeth Sztul
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35924
| |
Collapse
|
10
|
Jeng CJ, Fu SJ, You CY, Peng YJ, Hsiao CT, Chen TY, Tang CY. Defective Gating and Proteostasis of Human ClC-1 Chloride Channel: Molecular Pathophysiology of Myotonia Congenita. Front Neurol 2020; 11:76. [PMID: 32117034 PMCID: PMC7026490 DOI: 10.3389/fneur.2020.00076] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/22/2020] [Indexed: 01/17/2023] Open
Abstract
The voltage-dependent ClC-1 chloride channel, whose open probability increases with membrane potential depolarization, belongs to the superfamily of CLC channels/transporters. ClC-1 is almost exclusively expressed in skeletal muscles and is essential for stabilizing the excitability of muscle membranes. Elucidation of the molecular structures of human ClC-1 and several CLC homologs provides important insight to the gating and ion permeation mechanisms of this chloride channel. Mutations in the human CLCN1 gene, which encodes the ClC-1 channel, are associated with a hereditary skeletal muscle disease, myotonia congenita. Most disease-causing CLCN1 mutations lead to loss-of-function phenotypes in the ClC-1 channel and thus increase membrane excitability in skeletal muscles, consequently manifesting as delayed relaxations following voluntary muscle contractions in myotonic subjects. The inheritance pattern of myotonia congenita can be autosomal dominant (Thomsen type) or recessive (Becker type). To date over 200 myotonia-associated ClC-1 mutations have been identified, which are scattered throughout the entire protein sequence. The dominant inheritance pattern of some myotonia mutations may be explained by a dominant-negative effect on ClC-1 channel gating. For many other myotonia mutations, however, no clear relationship can be established between the inheritance pattern and the location of the mutation in the ClC-1 protein. Emerging evidence indicates that the effects of some mutations may entail impaired ClC-1 protein homeostasis (proteostasis). Proteostasis of membrane proteins comprises of biogenesis at the endoplasmic reticulum (ER), trafficking to the surface membrane, and protein turn-over at the plasma membrane. Maintenance of proteostasis requires the coordination of a wide variety of different molecular chaperones and protein quality control factors. A number of regulatory molecules have recently been shown to contribute to post-translational modifications of ClC-1 and play critical roles in the ER quality control, membrane trafficking, and peripheral quality control of this chloride channel. Further illumination of the mechanisms of ClC-1 proteostasis network will enhance our understanding of the molecular pathophysiology of myotonia congenita, and may also bring to light novel therapeutic targets for skeletal muscle dysfunction caused by myotonia and other pathological conditions.
Collapse
Affiliation(s)
- Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Ssu-Ju Fu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Ying You
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jheng Peng
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Tsung Hsiao
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Yu Chen
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,College of Medicine, Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Törnroth-Horsefield S. Phosphorylation of human AQP2 and its role in trafficking. VITAMINS AND HORMONES 2019; 112:95-117. [PMID: 32061351 DOI: 10.1016/bs.vh.2019.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human Aquaporin 2 (AQP2) is a membrane-bound water channel found in the kidney collecting duct whose regulation by trafficking plays a key role in regulating urine volume. AQP2 trafficking is tightly controlled by the pituitary hormone arginine vasopressin (AVP), which stimulates translocation of AQP2 residing in storage vesicles to the apical membrane. The AVP-dependent translocation of AQP2 to and from the apical membrane is controlled by multiple phosphorylation sites in the AQP2 C-terminus, the phosphorylation of which alters its affinity to proteins within the cellular membrane protein trafficking machinery. The aim of this chapter is to provide a summary of what is currently known about AVP-mediated AQP2 trafficking, dissecting the roles of individual phosphorylation sites, kinases and phosphatases and interacting proteins. From this, the picture of an immensely complex process emerges, of which many structural and molecular details remains to be elucidated.
Collapse
|
12
|
Kulkarni-Gosavi P, Makhoul C, Gleeson PA. Form and function of the Golgi apparatus: scaffolds, cytoskeleton and signalling. FEBS Lett 2019; 593:2289-2305. [PMID: 31378930 DOI: 10.1002/1873-3468.13567] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/09/2023]
Abstract
In addition to the classical functions of the Golgi in membrane transport and glycosylation, the Golgi apparatus of mammalian cells is now recognised to contribute to the regulation of a range of cellular processes, including mitosis, DNA repair, stress responses, autophagy, apoptosis and inflammation. These processes are often mediated, either directly or indirectly, by membrane scaffold molecules, such as golgins and GRASPs which are located on Golgi membranes. In many cases, these scaffold molecules also link the actin and microtubule cytoskeleton and influence Golgi morphology. An emerging theme is a strong relationship between the morphology of the Golgi and regulation of a variety of signalling pathways. Here, we review the molecular regulation of the morphology of the Golgi, especially the role of the golgins and other scaffolds in the interaction with the microtubule and actin networks. In addition, we discuss the impact of the modulation of the Golgi ribbon in various diseases, such as neurodegeneration and cancer, to the pathology of disease.
Collapse
Affiliation(s)
- Prajakta Kulkarni-Gosavi
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| | - Christian Makhoul
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| |
Collapse
|
13
|
Cargo Sorting at the trans-Golgi Network for Shunting into Specific Transport Routes: Role of Arf Small G Proteins and Adaptor Complexes. Cells 2019; 8:cells8060531. [PMID: 31163688 PMCID: PMC6627992 DOI: 10.3390/cells8060531] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/27/2023] Open
Abstract
The trans-Golgi network (TGN) is responsible for selectively recruiting newly synthesized cargo into transport carriers for delivery to their appropriate destination. In addition, the TGN is responsible for receiving and recycling cargo from endosomes. The membrane organization of the TGN facilitates the sorting of cargoes into distinct populations of transport vesicles. There have been significant advances in defining the molecular mechanism involved in the recognition of membrane cargoes for recruitment into different populations of transport carriers. This machinery includes cargo adaptors of the adaptor protein (AP) complex family, and monomeric Golgi-localized γ ear-containing Arf-binding protein (GGA) family, small G proteins, coat proteins, as well as accessory factors to promote budding and fission of transport vesicles. Here, we review this literature with a particular focus on the transport pathway(s) mediated by the individual cargo adaptors and the cargo motifs recognized by these adaptors. Defects in these cargo adaptors lead to a wide variety of diseases.
Collapse
|
14
|
Cavallini C, Zannini C, Olivi E, Tassinari R, Taglioli V, Rossi M, Poggi P, Chatgilialoglu A, Simonazzi G, Alviano F, Bonsi L, Ventura C. Restoring In Vivo-Like Membrane Lipidomics Promotes Exosome Trophic Behavior from Human Placental Mesenchymal Stromal/Stem Cells. Cell Transplant 2019; 27:55-69. [PMID: 29562775 PMCID: PMC6434476 DOI: 10.1177/0963689717723016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an effective tool in regenerative medicine notably for their intrinsic plentiful paracrine activity rather than differentiating properties. The hMSC secretome includes a wide spectrum of regulatory and trophic factors, encompassing several naked molecules as well as different kinds of extracellular vesicles (EVs). Among EVs, exosomes represent an intriguing population, able to shuttle proteins, transcription factors, and genetic materials, with a relevant role in cell-to-cell communication, modulating biological responses in recipient cells. In this context, the extracellular milieu can greatly impact the paracrine activity of stem cells, modifying their metabolism, and the dynamics of vesicle secretion. In the present study, we investigated the effects elicited on exosome patterning by tailored, ad hoc formulated lipid supplementation (Refeed®) in MSCs derived from human fetal membranes (hFM-MSCs). Wound healing experiments revealed that stem cell exposure to exosomes obtained from Refeed®-supplemented hFM-MSCs increased their migratory capability, although the amount of exosomes released after Refeed® supplementation was lower than that yielded from non-supplemented cells. We found that such a decrease was mainly due to a different rate of exosomal exocytosis rather than to an effect of the lipid supplement on the endocytic pathway. Endoplasmic reticulum homeostasis was modified by supplementation, through the upregulation of PKR-like ER kinase (PERK) and inositol-requiring enzyme 1α (IRE1α). Increased expression of these proteins did not lead to stress-induced, unfolded protein response (UPR)-mediated apoptosis, nor did it affect phosphorylation of p38 kinase, suggesting that PERK and IRE1α overexpression was due to augmented metabolic activities mediated by optimization of a cellular feeding network afforded through lipid supplementation. In summary, these results demonstrate how tailored lipid supplementation can successfully modify the paracrine features in hFM-MSCs, impacting both intracellular vesicle trafficking and secreted exosome number and function.
Collapse
Affiliation(s)
- Claudia Cavallini
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Chiara Zannini
- 3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy.,4 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Nephrology, Dialysis and Renal Transplant, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elena Olivi
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Riccardo Tassinari
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Valentina Taglioli
- 2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,6 Department of Experimental, Diagnostic and Specialty Medicine, Laboratory of Experimental Cardiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Martina Rossi
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | | | | | - Giuliana Simonazzi
- 8 Division of Obstetrics and Prenatal Medicine, Department of Medical and Surgical Sciences, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Francesco Alviano
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Laura Bonsi
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Carlo Ventura
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,9 CNR, Institute of Organic Synthesis and Photoreactivity (Istituto per la Sintesi Organica e la Fotoreattività ISOF), Via Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
15
|
Abstract
The Golgi apparatus is a central intracellular membrane-bound organelle with key functions in trafficking, processing, and sorting of newly synthesized membrane and secretory proteins and lipids. To best perform these functions, Golgi membranes form a unique stacked structure. The Golgi structure is dynamic but tightly regulated; it undergoes rapid disassembly and reassembly during the cell cycle of mammalian cells and is disrupted under certain stress and pathological conditions. In the past decade, significant amount of effort has been made to reveal the molecular mechanisms that regulate the Golgi membrane architecture and function. Here we review the major discoveries in the mechanisms of Golgi structure formation, regulation, and alteration in relation to its functions in physiological and pathological conditions to further our understanding of Golgi structure and function in health and diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Santamarï A-Gï Mez J, Mariscal V, Luque I. Mechanisms for Protein Redistribution in Thylakoids of Anabaena During Cell Differentiation. PLANT & CELL PHYSIOLOGY 2018; 59:1860-1873. [PMID: 29878163 DOI: 10.1093/pcp/pcy103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/25/2018] [Indexed: 06/08/2023]
Abstract
Thylakoid membranes are far from being homogeneous in composition. On the contrary, compositional heterogeneity of lipid and protein content is well known to exist in these membranes. The mechanisms for the confinement of proteins at a particular membrane domain have started to be unveiled, but we are far from a thorough understanding, and many issues remain to be elucidated. During the differentiation of heterocysts in filamentous cyanobacteria of the Anabaena and Nostoc genera, thylakoids undergo a complete reorganization, separating into two membrane domains of different appearance and subcellular localization. Evidence also indicates different functionality and protein composition for these two membrane domains. In this work, we have addressed the mechanisms that govern the specific localization of proteins at a particular membrane domain. Two classes of proteins were distinguished according to their distribution in the thylakoids. Our results indicate that the specific accumulation of proteins of the CURVATURE THYLAKOID 1 (CURT1) family and proteins containing the homologous CAAD domain at subpolar honeycomb thylakoids is mediated by multiple mechanisms including a previously unnoticed phenomenon of thylakoid membrane migration.
Collapse
Affiliation(s)
- Javier Santamarï A-Gï Mez
- Instituto de Bioqu�mica Vegetal y Fotos�ntesis, CSIC and Universidad de Sevilla, Avda Am�rico Vespucio 49, Seville E-41092, Spain
| | - Vicente Mariscal
- Instituto de Bioqu�mica Vegetal y Fotos�ntesis, CSIC and Universidad de Sevilla, Avda Am�rico Vespucio 49, Seville E-41092, Spain
| | - Ignacio Luque
- Instituto de Bioqu�mica Vegetal y Fotos�ntesis, CSIC and Universidad de Sevilla, Avda Am�rico Vespucio 49, Seville E-41092, Spain
| |
Collapse
|
17
|
Hsu RM, Zhong CY, Wang CL, Liao WC, Yang C, Lin SY, Lin JW, Cheng HY, Li PY, Yu CJ. Golgi tethering factor golgin-97 suppresses breast cancer cell invasiveness by modulating NF-κB activity. Cell Commun Signal 2018; 16:19. [PMID: 29703230 PMCID: PMC5923015 DOI: 10.1186/s12964-018-0230-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Background Golgin-97 is a tethering factor in the trans-Golgi network (TGN) and is crucial for vesicular trafficking and maintaining cell polarity. However, the significance of golgin-97 in human diseases such as cancer remains unclear. Methods We searched for a potential role of golgin-97 in cancers using Kaplan-Meier Plotter (http://kmplot.com) and Oncomine (www.oncomine.org) datasets. Specific functions of golgin-97 in migration and invasion were examined in golgin-97-knockdown and golgin-97-overexpressing cells. cDNA microarray, pathway analysis and qPCR were used to identify gene profiles regulated by golgin-97. The role of golgin-97 in NF-κB signaling pathway was examined by using subcellular fractionation, luciferase reporter assay, western blot analysis and immunofluorescence assay (IFA). Results We found that low expression of golgin-97 correlated with poor overall survival of cancer patients and was associated with invasiveness in breast cancer cells. Golgin-97 knockdown promoted cell migration and invasion, whereas re-expression of golgin-97 restored the above phenotypes in breast cancer cells. Microarray and pathway analyses revealed that golgin-97 knockdown induced the expression of several invasion-promoting genes that were transcriptionally regulated by NF-κB p65. Mechanistically, golgin-97 knockdown significantly reduced IκBα protein levels and activated NF-κB, whereas neither IκBα levels nor NF-κB activity was changed in TGN46- or GCC185-knockdown cells. Conversely, golgin-97 overexpression suppressed NF-κB activity and restored the levels of IκBα in golgin-97-knockdown cells. Interestingly, the results of Golgi-disturbing agent treatment revealed that the loss of Golgi integrity was not involved in the NF-κB activation induced by golgin-97 knockdown. Moreover, both TGN-bound and cytosolic golgin-97 inhibited NF-κB activation, indicating that golgin-97 functions as an NF-κB suppressor regardless of its subcellular localization. Conclusion Our results collectively demonstrate a novel and suppressive role of golgin-97 in cancer invasiveness. We also provide a new avenue for exploring the relationship between the TGN, golgin-97 and NF-κB signaling in tumor progression. Electronic supplementary material The online version of this article (10.1186/s12964-018-0230-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rae-Mann Hsu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cai-Yan Zhong
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Liang Wang
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Wei-Chao Liao
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Center for General Education, Chang Gung University, Taoyuan, Taiwan
| | - Chi Yang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Yu Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-Wei Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Yun Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Yu Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Jung Yu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
18
|
Auclair N, Melbouci L, St-Pierre D, Levy E. Gastrointestinal factors regulating lipid droplet formation in the intestine. Exp Cell Res 2018; 363:1-14. [PMID: 29305172 DOI: 10.1016/j.yexcr.2017.12.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
Cytoplasmic lipid droplets (CLD) are considered as neutral lipid reservoirs, which protect cells from lipotoxicity. It became clear that these fascinating dynamic organelles play a role not only in energy storage and metabolism, but also in cellular lipid and protein handling, inter-organelle communication, and signaling among diverse functions. Their dysregulation is associated with multiple disorders, including obesity, liver steatosis and cardiovascular diseases. The central aim of this review is to highlight the link between intra-enterocyte CLD dynamics and the formation of chylomicrons, the main intestinal dietary lipid vehicle, after overviewing the morphology, molecular composition, biogenesis and functions of CLD.
Collapse
Affiliation(s)
- N Auclair
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5
| | - L Melbouci
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - D St-Pierre
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - E Levy
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5; Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada G1V 0A6.
| |
Collapse
|
19
|
Roche JV, Törnroth-Horsefield S. Aquaporin Protein-Protein Interactions. Int J Mol Sci 2017; 18:ijms18112255. [PMID: 29077056 PMCID: PMC5713225 DOI: 10.3390/ijms18112255] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 12/20/2022] Open
Abstract
Aquaporins are tetrameric membrane-bound channels that facilitate transport of water and other small solutes across cell membranes. In eukaryotes, they are frequently regulated by gating or trafficking, allowing for the cell to control membrane permeability in a specific manner. Protein–protein interactions play crucial roles in both regulatory processes and also mediate alternative functions such as cell adhesion. In this review, we summarize recent knowledge about aquaporin protein–protein interactions; dividing the interactions into three types: (1) interactions between aquaporin tetramers; (2) interactions between aquaporin monomers within a tetramer (hetero-tetramerization); and (3) transient interactions with regulatory proteins. We particularly focus on the structural aspects of the interactions, discussing the small differences within a conserved overall fold that allow for aquaporins to be differentially regulated in an organism-, tissue- and trigger-specific manner. A deep knowledge about these differences is needed to fully understand aquaporin function and regulation in many physiological processes, and may enable design of compounds targeting specific aquaporins for treatment of human disease.
Collapse
Affiliation(s)
- Jennifer Virginia Roche
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| |
Collapse
|
20
|
Roche JV, Survery S, Kreida S, Nesverova V, Ampah-Korsah H, Gourdon M, Deen PMT, Törnroth-Horsefield S. Phosphorylation of human aquaporin 2 (AQP2) allosterically controls its interaction with the lysosomal trafficking protein LIP5. J Biol Chem 2017; 292:14636-14648. [PMID: 28710278 DOI: 10.1074/jbc.m117.788364] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 07/07/2017] [Indexed: 12/21/2022] Open
Abstract
The interaction between the renal water channel aquaporin-2 (AQP2) and the lysosomal trafficking regulator-interacting protein LIP5 targets AQP2 to multivesicular bodies and facilitates lysosomal degradation. This interaction is part of a process that controls AQP2 apical membrane abundance in a vasopressin-dependent manner, allowing for urine volume adjustment. Vasopressin regulates phosphorylation at four sites within the AQP2 C terminus (Ser256, Ser261, Ser264, and Thr269), of which Ser256 is crucial and sufficient for AQP2 translocation from storage vesicles to the apical membrane. However, whether AQP2 phosphorylation modulates AQP2-LIP5 complex affinity is unknown. Here we used far-Western blot analysis and microscale thermophoresis to show that the AQP2 binds LIP5 in a phosphorylation-dependent manner. We constructed five phospho-mimicking mutants (S256E, S261E, S264E, T269E, and S256E/T269E) and a C-terminal truncation mutant (ΔP242) that lacked all phosphorylation sites but retained a previously suggested LIP5-binding site. CD spectroscopy indicated that wild-type AQP2 and the phospho-mimicking mutants had similar overall structure but displayed differences in melting temperatures possibly arising from C-terminal conformational changes. Non-phosphorylated AQP2 bound LIP5 with the highest affinity, whereas AQP2-ΔP242 had 20-fold lower affinity as determined by microscale thermophoresis. AQP2-S256E, S261E, T269E, and S256E/T269E all had reduced affinity. This effect was most prominent for AQP2-S256E, which fits well with its role in apical membrane targeting. AQP2-S264E had affinity similar to non-phosphorylated AQP2, possibly indicating a role in exosome excretion. Our data suggest that AQP2 phosphorylation allosterically controls its interaction with LIP5, illustrating how altered affinities to interacting proteins form the basis for regulation of AQP2 trafficking by post-translational modifications.
Collapse
Affiliation(s)
- Jennifer Virginia Roche
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Sabeen Survery
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Stefan Kreida
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Veronika Nesverova
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Henry Ampah-Korsah
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Maria Gourdon
- From the Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden and
| | - Peter M T Deen
- the Department of Physiology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | | |
Collapse
|
21
|
Dysregulation of intracellular trafficking and endosomal sorting in Alzheimer's disease: controversies and unanswered questions. Biochem J 2017; 473:1977-93. [PMID: 27407168 DOI: 10.1042/bcj20160147] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid plaques in the brain consisting of an aggregated form of amyloid β-peptide (Aβ) derived from sequential amyloidogenic processing of the amyloid precursor protein (APP) by membrane-bound proteases β-site APP-cleaving enzyme 1 (BACE1) and γ-secretase. The initial processing of APP by BACE1 is re-gulated by intracellular sorting events of the enzyme, which is a prime target for therapeutic intervention. GWAS (genome-wide sequencing studies) have identified several AD-susceptibility genes that are associated with the regulation of membrane trafficking, and substantial evidence now indicates that AD is likely to arise from defective membrane trafficking in either or both of the secretory and endocytic pathways. Considerable progress has been made in defining the intracellular trafficking pathways of BACE1 and APP and the sorting signals of these membrane proteins that define their itineraries. In this review we highlight recent advances in understanding the regulation of the intracellular sorting of BACE1 and APP, discuss how dysregulation of these trafficking events may lead to enhanced generation of the neurotoxic Aβ products in AD and highlight the unresolved questions in the field.
Collapse
|
22
|
Triacca V, Güç E, Kilarski WW, Pisano M, Swartz MA. Transcellular Pathways in Lymphatic Endothelial Cells Regulate Changes in Solute Transport by Fluid Stress. Circ Res 2017; 120:1440-1452. [DOI: 10.1161/circresaha.116.309828] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/12/2023]
Abstract
Rationale:
The transport of interstitial fluid and solutes into lymphatic vessels is important for maintaining interstitial homeostasis and delivering antigens and soluble factors to the lymph node for immune surveillance. Transendothelial transport across lymphatic endothelial cells (LECs) is commonly considered to occur paracellularly, or between cell–cell junctions, and driven by local pressure and concentration gradients. However, emerging evidence suggests that LECs also play active roles in regulating interstitial solute balance and can scavenge and store antigens, raising the possibility that vesicular or transcellular pathways may be important in lymphatic solute transport.
Objective:
The aim of this study was to determine the relative importance of transcellular (vesicular) versus paracellular transport pathways by LECs and how mechanical stress (ie, fluid flow conditioning) alters either pathway.
Methods and Results:
We demonstrate that transcellular transport mechanisms substantially contribute to lymphatic solute transport and that solute uptake occurs in both caveolae- and clathrin-coated vesicles. In vivo, intracelluar uptake of fluorescently labeled albumin after intradermal injection by LECs was similar to that of dermal dendritic cells. In vitro, we developed a method to differentially quantify intracellular solute uptake versus transendothelial transport by LECs. LECs preconditioned to 1 µm/s transmural flow demonstrated increased uptake and basal-to-apical solute transport, which could be substantially reversed by blocking dynamin-dependent vesicle formation.
Conclusions:
These findings reveal the importance of intracellular transport in steady-state lymph formation and suggest that LECs use transcellular mechanisms in parallel to the well-described paracellular route to modulate solute transport from the interstitium according to biomechanical cues.
Collapse
Affiliation(s)
- Valentina Triacca
- From the Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (V.T., E.G., W.W.K., M.P., M.A.S.); and Institute for Molecular Engineering, The University of Chicago, IL (W.W.K., M.A.S.)
| | - Esra Güç
- From the Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (V.T., E.G., W.W.K., M.P., M.A.S.); and Institute for Molecular Engineering, The University of Chicago, IL (W.W.K., M.A.S.)
| | - Witold W. Kilarski
- From the Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (V.T., E.G., W.W.K., M.P., M.A.S.); and Institute for Molecular Engineering, The University of Chicago, IL (W.W.K., M.A.S.)
| | - Marco Pisano
- From the Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (V.T., E.G., W.W.K., M.P., M.A.S.); and Institute for Molecular Engineering, The University of Chicago, IL (W.W.K., M.A.S.)
| | - Melody A. Swartz
- From the Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (V.T., E.G., W.W.K., M.P., M.A.S.); and Institute for Molecular Engineering, The University of Chicago, IL (W.W.K., M.A.S.)
| |
Collapse
|
23
|
Murray M, Zhou F. Trafficking and other regulatory mechanisms for organic anion transporting polypeptides and organic anion transporters that modulate cellular drug and xenobiotic influx and that are dysregulated in disease. Br J Pharmacol 2017; 174:1908-1924. [PMID: 28299773 DOI: 10.1111/bph.13785] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 12/25/2022] Open
Abstract
Organic anion transporters (OATs) and organic anion-transporting polypeptides (OATPs), encoded by a number of solute carrier (SLC)22A and SLC organic anion (SLCO) genes, mediate the absorption and distribution of drugs and other xenobiotics. The regulation of OATs and OATPs is complex, comprising both transcriptional and post-translational mechanisms. Plasma membrane expression is required for cellular substrate influx by OATs/OATPs. Thus, interest in post-translational regulatory processes, including membrane targeting, endocytosis, recycling and degradation of transporter proteins, is increasing because these are critical for plasma membrane expression. After being synthesized, transporters undergo N-glycosylation in the endoplasmic reticulum and Golgi apparatus and are delivered to the plasma membrane by vesicular transport. Their expression at the cell surface is maintained by de novo synthesis and recycling, which occurs after clathrin- and/or caveolin-dependent endocytosis of existing protein. Several studies have shown that phosphorylation by signalling kinases is important for the internalization and recycling processes, although the transporter protein does not appear to be directly phosphorylated. After internalization, transporters that are targeted for degradation undergo ubiquitination, most likely on intracellular loop residues. Epigenetic mechanisms, including methylation of gene regulatory regions and transcription from alternate promoters, are also significant in the regulation of certain SLC22A/SLCO genes. The membrane expression of OATs/OATPs is dysregulated in disease, which affects drug efficacy and detoxification. Several transporters are expressed in the cytoplasmic subcompartment in disease states, which suggests that membrane targeting/internalization/recycling may be impaired. This article focuses on recent developments in OAT and OATP regulation, their dysregulation in disease and the significance for drug therapy.
Collapse
Affiliation(s)
- Michael Murray
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, NSW, 2006, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
24
|
Luo S, Jing L, Zhao T, Li Y, Liu Z, Diao A. Ubiquitination and dynactin regulate TMEPAI lysosomal trafficking. Sci Rep 2017; 7:42668. [PMID: 28218281 PMCID: PMC5316989 DOI: 10.1038/srep42668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/13/2017] [Indexed: 12/17/2022] Open
Abstract
The transmembrane prostate androgen-induced protein (TMEPAI) has been reported to be elevated in various tumor cells, is localized to the lysosome and promotes lysosome stability. The molecular mechanism of TMEPAI trafficking however to the lysosome is unknown. Here we report that clathrin and CI-M6PR mediate TMEPAI transport from the Golgi directly into the endo-lysosomal pathway. TMEPAI is ubiquitinated at its C-terminal region and ubiquitination modification of TMEPAI is a signal for its lysosomal trafficking. Moreover, TMEPAI binds the ubiquitin binding proteins Hrs and STAM which is required for its lysosomal transport. In addition, TMEPAI interacts with the dynactin pointed-end complex subunits dynactin 5 and dynactin 6. The aa 132–155 domain is essential for specific TMEPAI binding and deletion of this binding site leads to mis-trafficking of TMEPAI to the plasma membrane. These results reveal the pathway and mechanism regulating transport of TMEPAI to the lysosome, which helps to further understand the role of TMEPAI in tumorigenesis.
Collapse
Affiliation(s)
- Shenheng Luo
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| | - Lei Jing
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| | - Tian Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| | - Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| | - Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, Tianjin, 300457, China
| |
Collapse
|
25
|
Bhide GP, Colley KJ. Sialylation of N-glycans: mechanism, cellular compartmentalization and function. Histochem Cell Biol 2017; 147:149-174. [PMID: 27975143 PMCID: PMC7088086 DOI: 10.1007/s00418-016-1520-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2016] [Indexed: 12/18/2022]
Abstract
Sialylated N-glycans play essential roles in the immune system, pathogen recognition and cancer. This review approaches the sialylation of N-glycans from three perspectives. The first section focuses on the sialyltransferases that add sialic acid to N-glycans. Included in the discussion is a description of these enzymes' glycan acceptors, conserved domain organization and sequences, molecular structure and catalytic mechanism. In addition, we discuss the protein interactions underlying the polysialylation of a select group of adhesion and signaling molecules. In the second section, the biosynthesis of sialic acid, CMP-sialic acid and sialylated N-glycans is discussed, with a special emphasis on the compartmentalization of these processes in the mammalian cell. The sequences and mechanisms maintaining the sialyltransferases and other glycosylation enzymes in the Golgi are also reviewed. In the final section, we have chosen to discuss processes in which sialylated glycans, both N- and O-linked, play a role. The first part of this section focuses on sialic acid-binding proteins including viral hemagglutinins, Siglecs and selectins. In the second half of this section, we comment on the role of sialylated N-glycans in cancer, including the roles of β1-integrin and Fas receptor N-glycan sialylation in cancer cell survival and drug resistance, and the role of these sialylated proteins and polysialic acid in cancer metastasis.
Collapse
Affiliation(s)
- Gaurang P Bhide
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 S. Ashland Avenue, MC669, Chicago, IL, 60607, USA
| | - Karen J Colley
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 S. Ashland Avenue, MC669, Chicago, IL, 60607, USA.
| |
Collapse
|
26
|
Diseases of the Synaptic Vesicle: A Potential New Group of Neurometabolic Disorders Affecting Neurotransmission. Semin Pediatr Neurol 2016; 23:306-320. [PMID: 28284392 DOI: 10.1016/j.spen.2016.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The general concept of inborn error of metabolism is currently evolving into the interface between classical biochemistry and cellular biology. Basic neuroscience is providing increasing knowledge about the mechanisms of neurotransmission and novel related disorders are being described. There is a necessity of updating the classic concept of "inborn error of neurotransmitters (NT)" that considers mainly defects of synthesis and catabolism and transport of low weight NT molecules. Monogenic defects of the synaptic vesicle (SV), and especially those affecting the SV cycle are a potential new group of NT disorders since they end up in abnormal NT turnover and release. The most common clinical manifestations include epilepsy, intellectual disability, autism and movement disorders, and are in the continuum symptoms of synaptopathies. Interestingly, brain malformations and neurodegenerative conditions are also present within SV diseases. Metabolomics, proteomics, and other -omic techniques probably will provide biomarkers and contribute to therapeutic targets in the future.
Collapse
|
27
|
Blazek AD, Paleo BJ, Weisleder N. Plasma Membrane Repair: A Central Process for Maintaining Cellular Homeostasis. Physiology (Bethesda) 2016; 30:438-48. [PMID: 26525343 DOI: 10.1152/physiol.00019.2015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane repair is a conserved cellular response mediating active resealing of membrane disruptions to maintain homeostasis and prevent cell death and progression of multiple diseases. Cell membrane repair repurposes mechanisms from various cellular functions, including vesicle trafficking, exocytosis, and endocytosis, to mend the broken membrane. Recent studies increased our understanding of membrane repair by establishing the molecular machinery contributing to membrane resealing. Here, we review some of the key proteins linked to cell membrane repair.
Collapse
Affiliation(s)
- Alisa D Blazek
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Brian J Paleo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
28
|
Chantreau V, Taddese B, Munier M, Gourdin L, Henrion D, Rodien P, Chabbert M. Molecular Insights into the Transmembrane Domain of the Thyrotropin Receptor. PLoS One 2015; 10:e0142250. [PMID: 26545118 PMCID: PMC4636318 DOI: 10.1371/journal.pone.0142250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
The thyrotropin receptor (TSHR) is a G protein-coupled receptor (GPCR) that is member of the leucine-rich repeat subfamily (LGR). In the absence of crystal structure, the success of rational design of ligands targeting the receptor internal cavity depends on the quality of the TSHR models built. In this subfamily, transmembrane helices (TM) 2 and 5 are characterized by the absence of proline compared to most receptors, raising the question of the structural conformation of these helices. To gain insight into the structural properties of these helices, we carried out bioinformatics and experimental studies. Evolutionary analysis of the LGR family revealed a deletion in TM5 but provided no information on TM2. Wild type residues at positions 2.58, 2.59 or 2.60 in TM2 and/or at position 5.50 in TM5 were substituted to proline. Depending on the position of the proline substitution, different effects were observed on membrane expression, glycosylation, constitutive cAMP activity and responses to thyrotropin. Only proline substitution at position 2.59 maintained complex glycosylation and high membrane expression, supporting occurrence of a bulged TM2. The TSHR transmembrane domain was modeled by homology with the orexin 2 receptor, using a protocol that forced the deletion of one residue in the TM5 bulge of the template. The stability of the model was assessed by molecular dynamics simulations. TM5 straightened during the equilibration phase and was stable for the remainder of the simulations. Our data support a structural model of the TSHR transmembrane domain with a bulged TM2 and a straight TM5 that is specific of glycoprotein hormone receptors.
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acid Substitution
- Computational Biology
- Cyclic AMP/metabolism
- Evolution, Molecular
- Glycosylation
- HEK293 Cells
- Humans
- Models, Molecular
- Molecular Dynamics Simulation
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Phylogeny
- Protein Structure, Tertiary
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/classification
- Receptors, G-Protein-Coupled/genetics
- Receptors, Thyrotropin/chemistry
- Receptors, Thyrotropin/genetics
- Receptors, Thyrotropin/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sequence Deletion
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Vanessa Chantreau
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
| | - Bruck Taddese
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
| | - Mathilde Munier
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
| | - Louis Gourdin
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
- Reference Centre for the pathologies of hormonal receptivity, Department of Endocrinology, Centre Hospitalier Universitaire of Angers, Angers, France
| | - Daniel Henrion
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
| | - Patrice Rodien
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
- Reference Centre for the pathologies of hormonal receptivity, Department of Endocrinology, Centre Hospitalier Universitaire of Angers, Angers, France
| | - Marie Chabbert
- UMR CNRS 6214 –INSERM 1083, Laboratory of Integrated Neurovascular and Mitochondrial Biology, University of Angers, Angers, France
| |
Collapse
|
29
|
Schuberth CE, Tängemo C, Coneva C, Tischer C, Pepperkok R. Self-organization of core Golgi material is independent of COPII-mediated endoplasmic reticulum export. J Cell Sci 2015; 128:1279-93. [PMID: 25717003 DOI: 10.1242/jcs.154443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Golgi is a highly organized and dynamic organelle that receives and distributes material from and to the endoplasmic reticulum (ER) and the endocytic pathway. One open question about Golgi organization is whether it is solely based on ER-to-Golgi transport. Here, we analyzed the kinetics of Golgi breakdown in the absence of COPII-dependent ER export with high temporal and spatial resolution using quantitative fluorescence microscopy. We found that Golgi breakdown occurred in two phases. While Golgi enzymes continuously redistributed to the ER, we consistently observed extensive Golgi fragmentation at the beginning of the breakdown, followed by microtubule-dependent formation of a Golgi remnant structure (phase 1). Further Golgi disintegration occurred less uniformly (phase 2). Remarkably, cisternal Golgi morphology was lost early in phase 1 and Golgi fragments instead corresponded to variably sized vesicle clusters. These breakdown intermediates were devoid of COPI-dependent recycling material, but contained typical 'core' Golgi components. Furthermore, Golgi breakdown intermediates were able to disassemble and reassemble following cell division, indicating that they retained important regulatory capabilities. Taken together, these findings support the view that Golgi self-organization exists independently of ER-to-Golgi transport.
Collapse
Affiliation(s)
- Christian E Schuberth
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany Institute of Cell Dynamics and Imaging, University of Muenster, von-Esmarch-Str. 56, 48149 Muenster, Germany Cells in Motion Cluster of Excellence (EXC1003-CiM), University of Muenster, von-Esmarch-Str. 56, 48149 Muenster, Germany
| | - Carolina Tängemo
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Cvetalina Coneva
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Christian Tischer
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| |
Collapse
|
30
|
Abstract
Membrane trafficking depends on transport vesicles and carriers docking and fusing with the target organelle for the delivery of cargo. Membrane tethers and small guanosine triphosphatases (GTPases) mediate the docking of transport vesicles/carriers to enhance the efficiency of the subsequent SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-mediated fusion event with the target membrane bilayer. Different classes of membrane tethers and their specific intracellular location throughout the endomembrane system are now well defined. Recent biochemical and structural studies have led to a deeper understanding of the mechanism by which membrane tethers mediate docking of membrane carriers as well as an appreciation of the role of tethers in coordinating the correct SNARE complex and in regulating the organization of membrane compartments. This review will summarize the properties and roles of membrane tethers of both secretory and endocytic systems.
Collapse
Affiliation(s)
- Pei Zhi Cheryl Chia
- National Institute of Dental and Craniofacial Research, National Institutes of Health30 Convent Drive, Bethesda, MD 20892-4340USA
| | - Paul A. Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute30 Flemington Road, The University of Melbourne, Victoria 3010Australia
| |
Collapse
|
31
|
Nastou KC, Tsaousis GN, Kremizas KE, Litou ZI, Hamodrakas SJ. The human plasma membrane peripherome: visualization and analysis of interactions. BIOMED RESEARCH INTERNATIONAL 2014; 2014:397145. [PMID: 25057483 PMCID: PMC4095733 DOI: 10.1155/2014/397145] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/04/2014] [Indexed: 12/11/2022]
Abstract
A major part of membrane function is conducted by proteins, both integral and peripheral. Peripheral membrane proteins temporarily adhere to biological membranes, either to the lipid bilayer or to integral membrane proteins with noncovalent interactions. The aim of this study was to construct and analyze the interactions of the human plasma membrane peripheral proteins (peripherome hereinafter). For this purpose, we collected a dataset of peripheral proteins of the human plasma membrane. We also collected a dataset of experimentally verified interactions for these proteins. The interaction network created from this dataset has been visualized using Cytoscape. We grouped the proteins based on their subcellular location and clustered them using the MCL algorithm in order to detect functional modules. Moreover, functional and graph theory based analyses have been performed to assess biological features of the network. Interaction data with drug molecules show that ~10% of peripheral membrane proteins are targets for approved drugs, suggesting their potential implications in disease. In conclusion, we reveal novel features and properties regarding the protein-protein interaction network created by peripheral proteins of the human plasma membrane.
Collapse
Affiliation(s)
- Katerina C. Nastou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| | - Georgios N. Tsaousis
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| | - Kimon E. Kremizas
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| | - Zoi I. Litou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| | - Stavros J. Hamodrakas
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| |
Collapse
|
32
|
Van Roey K, Uyar B, Weatheritt RJ, Dinkel H, Seiler M, Budd A, Gibson TJ, Davey NE. Short Linear Motifs: Ubiquitous and Functionally Diverse Protein Interaction Modules Directing Cell Regulation. Chem Rev 2014; 114:6733-78. [DOI: 10.1021/cr400585q] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Kim Van Roey
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Bora Uyar
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Robert J. Weatheritt
- MRC
Laboratory of Molecular Biology (LMB), Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Holger Dinkel
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Markus Seiler
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Aidan Budd
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Toby J. Gibson
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Norman E. Davey
- Structural
and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Department
of Physiology, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
33
|
Heiker JT, Kunath A, Kosacka J, Flehmig G, Knigge A, Kern M, Stumvoll M, Kovacs P, Blüher M, Klöting N. Identification of genetic loci associated with different responses to high-fat diet-induced obesity in C57BL/6N and C57BL/6J substrains. Physiol Genomics 2014; 46:377-84. [DOI: 10.1152/physiolgenomics.00014.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We have recently demonstrated that C57BL/6NTac and C57BL/6JRj substrains are significantly different in their response to high-fat diet-induced obesity (DIO). The C57BL/6JRj substrain seems to be protected from DIO and genetic differences between C57BL/6J and C57BL/6N substrains at 11 single nucleotide polymorphism (SNP) loci have been identified. To define genetic variants as well as differences in parameters of glucose homeostasis and insulin sensitivity between C57BL/6NTac and C57BL/6JRj substrains that may explain the different response to DIO, we analyzed 208 first backcross (BC1) hybrids of C57BL/6NTac and C57BL/6JRj [(C57BL/6NTac × C57BL/6JRj)F1 × C57BL/6NTac] mice. Body weight, epigonadal and subcutaneous fat mass, circulating leptin, as well as parameters of glucose metabolism were measured after 10 wk of high-fat diet (HFD). Genetic profiling of BC1 hybrids were performed using TaqMan SNP genotyping assays. Furthermore, to assess whether SNP polymorphisms could affect mRNA level, we carried out gene expression analysis in murine liver samples. Human subcutaneous adipose tissue was used to verify murine data of SNAP29. We identified four sex-specific variants that are associated with the extent of HFD-induced weight gain and fat depot mass. BC1 hybrids carrying the combination of risk or beneficial alleles exhibit the phenotypical extremes of the parental strains. Murine and human SC expression analysis revealed Snap29 as strongest candidate. Our data indicate an important role of these loci in responsiveness to HFD-induced obesity and suggest genes of the synaptic vesicle release system such as Snap29 being involved in the regulation of high-fat DIO.
Collapse
Affiliation(s)
- John T. Heiker
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
- Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Anne Kunath
- IFB AdiposityDiseases, Junior Research Group 2 “Animal models of obesity”, Leipzig University, Leipzig, Germany; and
| | - Joanna Kosacka
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
| | - Gesine Flehmig
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
- IFB AdiposityDiseases, Leipzig University, Leipzig, Germany
| | - Anja Knigge
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
| | - Matthias Kern
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
- IFB AdiposityDiseases, Leipzig University, Leipzig, Germany
| | - Peter Kovacs
- IFB AdiposityDiseases, Leipzig University, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
- IFB AdiposityDiseases, Leipzig University, Leipzig, Germany
| | - Nora Klöting
- Department of Medicine, Endocrinology and Diabetes, Leipzig University, Leipzig, Germany
- IFB AdiposityDiseases, Junior Research Group 2 “Animal models of obesity”, Leipzig University, Leipzig, Germany; and
| |
Collapse
|
34
|
Chia PZC, Ramdzan YM, Houghton FJ, Hatters DM, Gleeson PA. High-throughput quantitation of intracellular trafficking and organelle disruption by flow cytometry. Traffic 2014; 15:572-82. [PMID: 24612275 DOI: 10.1111/tra.12161] [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] [Received: 11/25/2013] [Revised: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 12/15/2022]
Abstract
Current methods for the quantitation of membrane protein trafficking rely heavily on microscopy, which has limited quantitative capacity for analyses of cell populations and is cumbersome to perform. Here we describe a simple flow cytometry-based method that circumvents these limitations. The method utilizes fluorescent pulse-width measurements as a highly sensitive indicator to monitor the changes in intracellular distributions of a fluorescently labelled molecule in a cell. Pulse-width analysis enabled us to discriminate cells with target proteins in different intracellular locations including Golgi, lyso-endosomal network and the plasma membrane, as well as detecting morphological changes in organelles such as Golgi perturbation. The movement of endogenous and exogenous retrograde cargo was tracked from the plasma membrane-to-endosomes-to-Golgi, by decreasing pulse-width values. A block in transport upon RNAi-mediated ablation of transport machinery was readily quantified, demonstrating the versatility of this technique to identify pathway inhibitors. We also showed that pulse-width can be exploited to sort and recover cells based on different intracellular staining patterns, e.g. early endosomes and Golgi, opening up novel downstream applications. Overall, the method provides new capabilities for viewing membrane transport in thousands of cells per minute, unbiased analysis of the trafficking of cargo, and the potential for rapid screening of inhibitors of trafficking pathways.
Collapse
Affiliation(s)
- Pei Zhi Cheryl Chia
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | | | | | | | | |
Collapse
|
35
|
Lee S, Kim JI, Heo J, Lee I, Park S, Hwang MW, Bae JY, Park MS, Park HJ, Park MS. The anti-influenza virus effect of Phellinus igniarius extract. J Microbiol 2013; 51:676-81. [DOI: 10.1007/s12275-013-3384-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/22/2013] [Indexed: 11/28/2022]
|
36
|
Stow JL, Murray RZ. Intracellular trafficking and secretion of inflammatory cytokines. Cytokine Growth Factor Rev 2013; 24:227-39. [PMID: 23647915 DOI: 10.1016/j.cytogfr.2013.04.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The secretion of cytokines by immune cells plays a significant role in determining the course of an inflammatory response. The levels and timing of each cytokine released are critical for mounting an effective but confined response, whereas excessive or dysregulated inflammation contributes to many diseases. Cytokines are both culprits and targets for effective treatments in some diseases. The multiple points and mechanisms that have evolved for cellular control of cytokine secretion highlight the potency of these mediators and the fine tuning required to manage inflammation. Cytokine production in cells is regulated by cell signaling, and at mRNA and protein synthesis levels. Thereafter, the intracellular transport pathways and molecular trafficking machinery have intricate and essential roles in dictating the release and activity of cytokines. The trafficking machinery and secretory (exocytic) pathways are complex and highly regulated in many cells, involving specialized membranes, molecules and organelles that enable these cells to deliver cytokines to often-distinct areas of the cell surface, in a timely manner. This review provides an overview of secretory pathways - both conventional and unconventional - and key families of trafficking machinery. The prevailing knowledge about the trafficking and secretion of a number of individual cytokines is also summarized. In conclusion, we present emerging concepts about the functional plasticity of secretory pathways and their modulation for controlling cytokines and inflammation.
Collapse
Affiliation(s)
- Jennifer L Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| | | |
Collapse
|
37
|
Hong MJ, Lee YM, Son YS, Im CH, Yi YB, Rim YG, Bahk JD, Heo JB. Rice Rab11 is required for JA-mediated defense signaling. Biochem Biophys Res Commun 2013; 434:797-802. [DOI: 10.1016/j.bbrc.2013.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 04/11/2013] [Indexed: 10/26/2022]
|
38
|
Abstract
The Golgi complex is considered the central station of the secretory pathway where cargo proteins and lipids are properly modified, classified, packed into specific carriers and delivered to their final destinations. Early electron microscope studies showed the extraordinary structural complexity of this organelle. However, despite the large volume of incoming and outgoing traffic, it is able to maintain its architecture, although it is also flexible enough to adapt to the functional status of the cell. Many components of the molecular machinery involved in membrane traffic and other Golgi functions have been identified. However, some basic aspects of Golgi functioning remain unsolved. For instance, how cargo moves through the stack remains controversial and two classical models have been proposed: vesicular transport and cisternal maturation. Since neither of these models explains all the experimental data, a combination of these models as well as new models have been proposed. In this context, the specific role of the cisternae, vesicles and tubules needs to be clarified. In this review, we summarize our current knowledge of the Golgi organization and function, focusing on the mechanisms of intra-Golgi transport.
Collapse
|
39
|
Abstract
LDs (lipid droplets) carrying TAG (triacylglycerol) and cholesteryl esters are emerging as dynamic cellular organelles that are generated in nearly every cell. They play a key role in lipid and membrane homoeostasis. Abnormal LD dynamics are associated with the pathophysiology of many metabolic diseases, such as obesity, diabetes, atherosclerosis, fatty liver and even cancer. Chylomicrons, stable droplets also consisting of TAG and cholesterol are generated in the intestinal epithelium to transport exogenous (dietary) lipids after meals from the small intestine to tissues for degradation. Defective chylomicron formation is responsible for inherited lipoprotein deficiencies, including abetalipoproteinaemia, hypobetalipoproteinaemia and chylomicron retention disease. These are disorders sharing characteristics such as fat malabsorption, low levels of circulating lipids and fat-soluble vitamins, failure to thrive in early childhood, ataxic neuropathy and visual impairment. Thus understanding the molecular mechanisms governing the dynamics of LDs and chylomicrons, namely, their biogenesis, growth, maintenance and degradation, will not only clarify their molecular role, but might also provide additional indications to treatment of metabolic diseases. In this review, we highlight the role of two small GTPases [ARFRP1 (ADP-ribosylation factor related protein 1) and ARL1 (ADP-ribosylation factor-like 1)] and their downstream targets acting on the trans-Golgi (Golgins and Rab proteins) on LD and chylomicron formation.
Collapse
|
40
|
Ng EL, Gan BQ, Ng F, Tang BL. Rab GTPases regulating receptor trafficking at the late endosome-lysosome membranes. Cell Biochem Funct 2012; 30:515-23. [DOI: 10.1002/cbf.2827] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 02/07/2012] [Accepted: 03/09/2012] [Indexed: 02/05/2023]
Affiliation(s)
- Ee Ling Ng
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Bin Qi Gan
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Fanny Ng
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Bor Luen Tang
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| |
Collapse
|
41
|
Brunholz S, Sisodia S, Lorenzo A, Deyts C, Kins S, Morfini G. Axonal transport of APP and the spatial regulation of APP cleavage and function in neuronal cells. Exp Brain Res 2012; 217:353-64. [PMID: 21960299 PMCID: PMC3670699 DOI: 10.1007/s00221-011-2870-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 09/07/2011] [Indexed: 12/12/2022]
Abstract
Over two decades have passed since the original discovery of amyloid precursor protein (APP). While physiological function(s) of APP still remain a matter of debate, consensus exists that the proteolytic processing of this protein represents a critical event in the life of neurons and that abnormalities in this process are instrumental in Alzheimer's disease (AD) pathogenesis. Specific molecular components involved in APP proteolysis have been identified, and their enzymatic activities characterized in great detail. As specific proteolytic fragments of APP are identified and novel physiological effects for these fragments are revealed, more obvious becomes our need to understand the spatial organization of APP proteolysis. Valuable insights on this process have been obtained through the study of non-neuronal cells. However, much less is known about the topology of APP processing in neuronal cells, which are characterized by their remarkably complex cellular architecture and extreme degree of polarization. In this review, we discuss published literature addressing various molecular mechanisms and components involved in the trafficking and subcellular distribution of APP and APP secretases in neurons. These include the relevant machinery involved in their sorting, the identity of membranous organelles in which APP is transported, and the molecular motor-based mechanisms involved in their translocation. We also review experimental evidence specifically addressing the processing of APP at the axonal compartment. Understanding neuron-specific mechanisms of APP processing would help illuminating the physiological roles of APP-derived proteolytic fragments and provide novel insights on AD pathogenesis.
Collapse
Affiliation(s)
- Silke Brunholz
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Houghton FJ, Bellingham SA, Hill AF, Bourges D, Ang DK, Gemetzis T, Gasnereau I, Gleeson PA. Arl5b is a Golgi-localised small G protein involved in the regulation of retrograde transport. Exp Cell Res 2012; 318:464-77. [DOI: 10.1016/j.yexcr.2011.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 12/04/2011] [Accepted: 12/28/2011] [Indexed: 11/30/2022]
|
43
|
Abstract
Protein traffic is necessary to maintain homeostasis in all eukaryotic organisms. All newly synthesized secretory proteins destined to the secretory and endolysosmal systems are transported from the endoplasmic reticulum to the Golgi before delivery to their final destinations. Here, we describe the COPII and COPI coating machineries that generate carrier vesicles and the tethers and SNAREs that mediate COPII and COPI vesicle fusion at the ER-Golgi interface.
Collapse
Affiliation(s)
- Tomasz Szul
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
44
|
Klann M, Koeppl H, Reuss M. Spatial modeling of vesicle transport and the cytoskeleton: the challenge of hitting the right road. PLoS One 2012; 7:e29645. [PMID: 22253752 PMCID: PMC3257240 DOI: 10.1371/journal.pone.0029645] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/02/2011] [Indexed: 01/15/2023] Open
Abstract
The membrane trafficking machinery provides a transport and sorting system for many cellular proteins. We propose a mechanistic agent-based computer simulation to integrate and test the hypothesis of vesicle transport embedded into a detailed model cell. The method tracks both the number and location of the vesicles. Thus both the stochastic properties due to the low numbers and the spatial aspects are preserved. The underlying molecular interactions that control the vesicle actions are included in a multi-scale manner based on the model of Heinrich and Rapoport (2005). By adding motor proteins we can improve the recycling process of SNAREs and model cell polarization. Our model also predicts that coat molecules should have a high turnover at the compartment membranes, while the turnover of motor proteins has to be slow. The modular structure of the underlying model keeps it tractable despite the overall complexity of the vesicle system. We apply our model to receptor-mediated endocytosis and show how a polarized cytoskeleton structure leads to polarized distributions in the plasma membrane both of SNAREs and the Ste2p receptor in yeast. In addition, we can couple signal transduction and membrane trafficking steps in one simulation, which enables analyzing the effect of receptor-mediated endocytosis on signaling.
Collapse
Affiliation(s)
- Michael Klann
- Automatic Control Laboratory, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland.
| | | | | |
Collapse
|
45
|
Lee HY, Bowen CH, Popescu GV, Kang HG, Kato N, Ma S, Dinesh-Kumar S, Snyder M, Popescu SC. Arabidopsis RTNLB1 and RTNLB2 Reticulon-like proteins regulate intracellular trafficking and activity of the FLS2 immune receptor. THE PLANT CELL 2011; 23:3374-91. [PMID: 21949153 PMCID: PMC3203430 DOI: 10.1105/tpc.111.089656] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 08/26/2011] [Accepted: 09/12/2011] [Indexed: 05/18/2023]
Abstract
Receptors localized at the plasma membrane are critical for the recognition of pathogens. The molecular determinants that regulate receptor transport to the plasma membrane are poorly understood. In a screen for proteins that interact with the FLAGELIN-SENSITIVE2 (FLS2) receptor using Arabidopsis thaliana protein microarrays, we identified the reticulon-like protein RTNLB1. We showed that FLS2 interacts in vivo with both RTNLB1 and its homolog RTNLB2 and that a Ser-rich region in the N-terminal tail of RTNLB1 is critical for the interaction with FLS2. Transgenic plants that lack RTNLB1 and RTNLB2 (rtnlb1 rtnlb2) or overexpress RTNLB1 (RTNLB1ox) exhibit reduced activation of FLS2-dependent signaling and increased susceptibility to pathogens. In both rtnlb1 rtnlb2 and RTNLB1ox, FLS2 accumulation at the plasma membrane was significantly affected compared with the wild type. Transient overexpression of RTNLB1 led to FLS2 retention in the endoplasmic reticulum (ER) and affected FLS2 glycosylation but not FLS2 stability. Removal of the critical N-terminal Ser-rich region or either of the two Tyr-dependent sorting motifs from RTNLB1 causes partial reversion of the negative effects of excess RTNLB1 on FLS2 transport out of the ER and accumulation at the membrane. The results are consistent with a model whereby RTNLB1 and RTNLB2 regulate the transport of newly synthesized FLS2 to the plasma membrane.
Collapse
Affiliation(s)
- Hyoung Yool Lee
- Boyce Thompson Institute for Plant Research, Ithaca, New York 14853
| | | | - George Viorel Popescu
- National Institute for Laser, Plasma, and Radiation Physics, Magurele 077125 Bucharest, Romania
| | - Hong-Gu Kang
- Boyce Thompson Institute for Plant Research, Ithaca, New York 14853
| | - Naohiro Kato
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803
| | - Shisong Ma
- College of Biological Sciences, University of California, Davis, California 95616
| | | | - Michael Snyder
- Department of Genetics, Stanford University, Stanford, California 94305
| | - Sorina Claudia Popescu
- Boyce Thompson Institute for Plant Research, Ithaca, New York 14853
- Address correspondence to
| |
Collapse
|
46
|
Zhi P, Chia PZC, Chia C, Gleeson PA. Intracellular trafficking of the β-secretase and processing of amyloid precursor protein. IUBMB Life 2011; 63:721-9. [PMID: 21834057 DOI: 10.1002/iub.512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/11/2011] [Indexed: 12/15/2022]
Abstract
The main component of the amyloid plaques found in the brains of those with Alzheimer's disease (AD) is a polymerized form of the β-amyloid peptide (Aβ) and is considered to play a central role in the pathogenesis of this neurodegenerative disorder. Aβ is derived from the proteolytic processing of the amyloid precursor protein (APP). Beta site APP-cleaving enzyme, BACE1 (also known as β-secretase) is a membrane-bound aspartyl protease responsible for the initial step in the generation of Aβ peptide and is thus a prime target for therapeutic intervention. Substantive evidence now indicates that the processing of APP by BACE1 is regulated by the intracellular sorting of the enzyme and, moreover, perturbations in these intracellular trafficking pathways have been linked to late-onset AD. In this review, we highlight the recent advances in the understanding of the regulation of the intracellular sorting of BACE1 and APP and illustrate why the trafficking of these cargos represent a key issue for understanding the membrane-mediated events associated with the generation of the neurotoxic Aβ products in AD.
Collapse
Affiliation(s)
- Pei Zhi
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
47
|
Abstract
Eukaryotic cells comprise a set of organelles, surrounded by membranes with a unique composition, which is maintained by a complex synthesis and transport system. Cells also synthesize the proteins destined for secretion. Together, these processes are known as the secretory pathway or exocytosis. In addition, many molecules can be internalized by cells through a process called endocytosis. Chronic and acute alcohol (ethanol) exposure alters the secretion of different essential products, such as hormones, neurotransmitters and others in a variety of cells, including central nervous system cells. This effect could be due to a range of mechanisms, including alcohol-induced alterations in the different steps involved in intracellular transport, such as glycosylation and vesicular transport along cytoskeleton elements. Moreover, alcohol consumption during pregnancy disrupts developmental processes in the central nervous system. No single mechanism has proved sufficient to account for these effects, and multiple factors are likely involved. One such mechanism indicates that ethanol also perturbs protein trafficking. The purpose of this review is to summarize our understanding of how ethanol exposure alters the trafficking of proteins in different cell systems, especially in central nervous system cells (neurons and astrocytes) in adult and developing brains.
Collapse
|
48
|
Benjamin JJR, Poon PP, Drysdale JD, Wang X, Singer RA, Johnston GC. Dysregulated Arl1, a regulator of post-Golgi vesicle tethering, can inhibit endosomal transport and cell proliferation in yeast. Mol Biol Cell 2011; 22:2337-47. [PMID: 21562219 PMCID: PMC3128535 DOI: 10.1091/mbc.e10-09-0765] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Small monomeric G proteins regulated in part by GTPase-activating proteins (GAPs) are molecular switches for several aspects of vesicular transport. The yeast Gcs1 protein is a dual-specificity GAP for ADP-ribosylation factor (Arf) and Arf-like (Arl)1 G proteins, and also has GAP-independent activities. The absence of Gcs1 imposes cold sensitivity for growth and endosomal transport; here we present evidence that dysregulated Arl1 may cause these impairments. We show that gene deletions affecting the Arl1 or Ypt6 vesicle-tethering pathways prevent Arl1 activation and membrane localization, and restore growth and trafficking in the absence of Gcs1. A mutant version of Gcs1 deficient for both ArfGAP and Arl1GAP activity in vitro still allows growth and endosomal transport, suggesting that the function of Gcs1 that is required for these processes is independent of GAP activity. We propose that, in the absence of this GAP-independent regulation by Gcs1, the resulting dysregulated Arl1 prevents growth and impairs endosomal transport at low temperatures. In cells with dysregulated Arl1, an increased abundance of the Arl1 effector Imh1 restores growth and trafficking, and does so through Arl1 binding. Protein sequestration at the trans-Golgi membrane by dysregulated, active Arl1 may therefore be the mechanism of inhibition.
Collapse
Affiliation(s)
- Jeremy J R Benjamin
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | | | | | | | |
Collapse
|
49
|
Wickström SA, Fässler R. Regulation of membrane traffic by integrin signaling. Trends Cell Biol 2011; 21:266-73. [DOI: 10.1016/j.tcb.2011.02.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 02/20/2011] [Accepted: 02/23/2011] [Indexed: 01/23/2023]
|
50
|
Abstract
The eukaryotic Golgi apparatus is characterized by a stack of flattened cisternae that are surrounded by transport vesicles. The organization and function of the Golgi require Golgi matrix proteins, including GRASPs and golgins, which exist primarily as fiber-like bridges between Golgi cisternae or between cisternae and vesicles. In this review, we highlight recent findings on Golgi matrix proteins, including their roles in maintaining the Golgi structure, vesicle tethering, and novel, unexpected functions. These new discoveries further our understanding of the molecular mechanisms that maintain the structure and the function of the Golgi, as well as its relationship with other cellular organelles such as the centrosome.
Collapse
|