1
|
Ono K, Niwa M, Suzuki H, Kobayashi NB, Yoshida T, Sawada M. Calmodulin as a Key Regulator of Exosomal Signal Peptides. Cells 2022; 12:cells12010158. [PMID: 36611951 PMCID: PMC9818429 DOI: 10.3390/cells12010158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Signal peptides (SPs) and their fragments play important roles as biomarkers and substances with physiological functions in extracellular fluid. We previously reported that SP fragments were released into extracellular fluid via exosomes and bound to calmodulin (CaM), an exosomal component, in a cell-free system. However, it currently remains unclear whether CaM intracellularly interacts with SP fragments or is involved in the trafficking of these fragments to exosomes. Therefore, the present study examined the binding of CaM to SP fragments in T-REx AspALP cells, transformed HEK293 cells expressing amyloid precursor protein (APP) SP flanking a reporter protein, and their exosomes. APP SP fragments were detected in exosomes from T-REx AspALP cells in the absence of W13, a CaM inhibitor, but were present in lower amounts in exosomes from W13-treated cells. Cargo proteins, such as Alix, CD63, and CD81, were increased in W13-treated T-REx AspALP cells but were decreased in their exosomes. Furthermore, CaM interacted with heat shock protein 70 and CD81 in T-REx AspALP cells and this increased in the presence of W13. APP SP fragments were detected in intracellular CaM complexes in the absence of W13, but not in its presence. These results indicate that CaM functions as a key regulator of the transport of SP fragments into exosomes and plays novel roles in the sorting of contents during exosomal biogenesis.
Collapse
Affiliation(s)
- Kenji Ono
- Department of Brain Function, Division of Stress Adaptation and Protection, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
- Department of Molecular Pharmacokinetics, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
- Correspondence: ; Tel.: +81-52-789-5002; Fax: +81-52-789-3994
| | - Mikio Niwa
- Institute for Advanced Sciences, Toagosei Co., Ltd., Tsukuba 300-2611, Ibaraki, Japan
| | - Hiromi Suzuki
- Department of Brain Function, Division of Stress Adaptation and Protection, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
- Department of Molecular Pharmacokinetics, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
| | | | - Tetsuhiko Yoshida
- Institute for Advanced Sciences, Toagosei Co., Ltd., Tsukuba 300-2611, Ibaraki, Japan
| | - Makoto Sawada
- Department of Brain Function, Division of Stress Adaptation and Protection, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
- Department of Molecular Pharmacokinetics, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Aichi, Japan
| |
Collapse
|
2
|
Golgi Metal Ion Homeostasis in Human Health and Diseases. Cells 2022; 11:cells11020289. [PMID: 35053405 PMCID: PMC8773785 DOI: 10.3390/cells11020289] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
The Golgi apparatus is a membrane organelle located in the center of the protein processing and trafficking pathway. It consists of sub-compartments with distinct biochemical compositions and functions. Main functions of the Golgi, including membrane trafficking, protein glycosylation, and sorting, require a well-maintained stable microenvironment in the sub-compartments of the Golgi, along with metal ion homeostasis. Metal ions, such as Ca2+, Mn2+, Zn2+, and Cu2+, are important cofactors of many Golgi resident glycosylation enzymes. The homeostasis of metal ions in the secretory pathway, which is required for proper function and stress response of the Golgi, is tightly regulated and maintained by transporters. Mutations in the transporters cause human diseases. Here we provide a review specifically focusing on the transporters that maintain Golgi metal ion homeostasis under physiological conditions and their alterations in diseases.
Collapse
|
3
|
Sontyana B, Shrivastava R, Battu S, Ghosh S, Mukhopadhyay S. Phagosome maturation and modulation of macrophage effector function by intracellular pathogens: target for therapeutics. Future Microbiol 2021; 17:59-76. [PMID: 34877879 DOI: 10.2217/fmb-2021-0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Macrophages are important cells that regulate various innate functions. Macrophages after engulfment of pathogens proceed for phagosome maturation and finally fuse with lysosomes to kill pathogens. Although pathogen degradation is one of the important functions of phagosomes, various immune-effector functions of macrophages are also dependent on the phagosome maturation process. This review discusses signaling processes regulating phagosome maturation as well as various effector functions of macrophages such as apoptosis, antigen presentation, autophagy and inflammasome that are dependent on the phagosome maturation process. It also discusses strategies adopted by various intracellular pathogens to counteract these functions to evade intracellular destruction mechanisms. These studies may give direction for the development of new therapeutics to control various intracellular infections.
Collapse
Affiliation(s)
- Brahmaji Sontyana
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rohini Shrivastava
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Srikanth Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| | - Sudip Ghosh
- Molecular Biology Unit, ICMR-National Institute of Nutrition, Jamai Osmania PO, Hyderabad, 500007, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| |
Collapse
|
4
|
Wenzhong L, Hualan L. COVID-19: the CaMKII-like system of S protein drives membrane fusion and induces syncytial multinucleated giant cells. Immunol Res 2021; 69:496-519. [PMID: 34410575 PMCID: PMC8374125 DOI: 10.1007/s12026-021-09224-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/24/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 S protein on the membrane of infected cells can promote receptor-dependent syncytia formation, relating to extensive tissue damage and lymphocyte elimination. In this case, it is challenging to obtain neutralizing antibodies and prevent them through antibodies effectively. Considering that, in the current study, structural domain search methods are adopted to analyze the SARS-CoV-2 S protein to find the fusion mechanism. The results show that after the EF-hand domain of S protein bound to calcium ions, S2 protein had CaMKII protein activities. Besides, the CaMKII_AD domain of S2 changed S2 conformation, facilitating the formation of HR1-HR2 six-helix bundles. Apart from that, the Ca2+-ATPase of S2 pumped calcium ions from the virus cytoplasm to help membrane fusion, while motor structures of S drove the CaATP_NAI and CaMKII_AD domains to extend to the outside and combined the viral membrane and the cell membrane, thus forming a calcium bridge. Furthermore, the phospholipid-flipping-ATPase released water, triggering lipid mixing and fusion and generating fusion pores. Then, motor structures promoted fusion pore extension, followed by the cytoplasmic contents of the virus being discharged into the cell cytoplasm. After that, the membrane of the virus slid onto the cell membrane along the flowing membrane on the gap of the three CaATP_NAI. At last, the HR1-HR2 hexamer would fall into the cytoplasm or stay on the cell membrane. Therefore, the CaMKII_like system of S protein facilitated membrane fusion for further inducing syncytial multinucleated giant cells.
Collapse
Affiliation(s)
- Liu Wenzhong
- School of Computer Science and Engineering, Sichuan University of Science & Engineering, Zigong, 643002, China.
- School of Life Science and Food Engineering, Yibin University, Yibin, 644000, China.
| | - Li Hualan
- School of Life Science and Food Engineering, Yibin University, Yibin, 644000, China
| |
Collapse
|
5
|
Flotillin-Dependent Membrane Microdomains Are Required for Functional Phagolysosomes against Fungal Infections. Cell Rep 2021; 32:108017. [PMID: 32814035 PMCID: PMC10054021 DOI: 10.1016/j.celrep.2020.108017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/03/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
Lipid rafts form signaling platforms on biological membranes with incompletely characterized role in immune response to infection. Here we report that lipid-raft microdomains are essential components of phagolysosomal membranes of macrophages and depend on flotillins. Genetic deletion of flotillins demonstrates that the assembly of both major defense complexes vATPase and NADPH oxidase requires membrane microdomains. Furthermore, we describe a virulence mechanism leading to dysregulation of membrane microdomains by melanized wild-type conidia of the important human-pathogenic fungus Aspergillus fumigatus resulting in reduced phagolysosomal acidification. We show that phagolysosomes with ingested melanized conidia contain a reduced amount of free Ca2+ ions and that inhibition of Ca2+-dependent calmodulin activity led to reduced lipid-raft formation. We identify a single-nucleotide polymorphism in the human FLOT1 gene resulting in heightened susceptibility for invasive aspergillosis in hematopoietic stem cell transplant recipients. Collectively, flotillin-dependent microdomains on the phagolysosomal membrane play an essential role in protective antifungal immunity.
Collapse
|
6
|
Veerana M, Mitra S, Ki S, Kim S, Choi E, Lee T, Park G. Plasma-mediated enhancement of enzyme secretion in Aspergillus oryzae. Microb Biotechnol 2021; 14:262-276. [PMID: 33151631 PMCID: PMC7888467 DOI: 10.1111/1751-7915.13696] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/08/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Technical bottlenecks in protein production and secretion often limit the efficient and robust industrial use of microbial enzymes. The potential of non-thermal atmospheric pressure plasma to overcome these technical barriers was examined. Spores of the fermenting fungus Aspergillus oryzae (A. oryzae) were submerged in potato dextrose broth (PDB) (5 × 106 per ml) and treated with micro dielectric barrier discharge plasma at an input voltage of 1.2 kV and current of 50 to 63 mA using nitrogen as the feed gas. The specific activity of α-amylase in the broth was increased by 7.4 to 9.3% after 24 and 48 h of plasma treatment. Long-lived species, such as NO2 - and NO3 - , generated in PDB after plasma treatment may have contributed to the elevated secretion of α-amylase. Observations after 24 h of plasma treatment also included increased accumulation of vesicles at the hyphal tip, hyphal membrane depolarization and higher intracellular Ca2+ levels. These results suggest that long-lived nitrogen species generated in PDB after plasma treatment can enhance the secretion of α-amylase from fungal hyphae by depolarizing the cell membrane and activating Ca2+ influx into hyphal cells, eventually leading to the accumulation of secretory vesicles near the hyphal tips.
Collapse
Affiliation(s)
- Mayura Veerana
- Department of Plasma Bioscience and DisplayKwangwoon UniversitySeoul01897Korea
| | - Sarmistha Mitra
- Department of Plasma Bioscience and DisplayKwangwoon UniversitySeoul01897Korea
| | - Se‐Hoon Ki
- Department of Electrical and Biological PhysicsKwangwoon UniversitySeoul01897Korea
| | - Soo‐Min Kim
- Department of Chemical EngineeringKwangwoon UniversitySeoul01897Korea
| | - Eun‐Ha Choi
- Department of Plasma Bioscience and DisplayKwangwoon UniversitySeoul01897Korea
- Department of Electrical and Biological PhysicsKwangwoon UniversitySeoul01897Korea
| | - Taek Lee
- Department of Chemical EngineeringKwangwoon UniversitySeoul01897Korea
| | - Gyungsoon Park
- Department of Plasma Bioscience and DisplayKwangwoon UniversitySeoul01897Korea
- Department of Electrical and Biological PhysicsKwangwoon UniversitySeoul01897Korea
| |
Collapse
|
7
|
Lloyd-Evans E, Waller-Evans H. Lysosomal Ca 2+ Homeostasis and Signaling in Health and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035311. [PMID: 31653642 DOI: 10.1101/cshperspect.a035311] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Calcium (Ca2+) signaling is an essential process in all cells that is maintained by a plethora of channels, pumps, transporters, receptors, and intracellular Ca2+ sequestering stores. Changes in cytosolic Ca2+ concentration govern processes as far reaching as fertilization, cell growth, and motility through to cell death. In recent years, lysosomes have emerged as a major intracellular Ca2+ storage organelle with an increasing involvement in triggering or regulating cellular functions such as endocytosis, autophagy, and Ca2+ release from the endoplasmic reticulum. This review will summarize recent work in the area of lysosomal Ca2+ signaling and homeostasis, including newly identified functions, and the involvement of lysosome-derived Ca2+ signals in human disease. In addition, we explore recent controversies in the techniques used for measurement of lysosomal Ca2+ content.
Collapse
Affiliation(s)
- Emyr Lloyd-Evans
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - Helen Waller-Evans
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, United Kingdom
| |
Collapse
|
8
|
Mukherjee K, Khatua B, Mandal C. Sialic Acid-Siglec-E Interactions During Pseudomonas aeruginosa Infection of Macrophages Interferes With Phagosome Maturation by Altering Intracellular Calcium Concentrations. Front Immunol 2020; 11:332. [PMID: 32184783 PMCID: PMC7059019 DOI: 10.3389/fimmu.2020.00332] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/10/2020] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is commonly associated with nosocomial and chronic infections of lungs. We have earlier demonstrated that an acidic sugar, sialic acid, is present in PA which is recognized and bound by sialic acid binding immunoglobulin type lectins (siglecs) expressed on neutrophils. Here, we have tried to gain a detailed insight into the immunosuppressive role of sialic acid-siglec interactions in macrophage-mediated clearance of sialylated PA (PA+Sia). We have demonstrated that PA+Sia shows enhanced binding (~1.5-fold) to macrophages due to additional interactions between sialic acids and siglec-E and exhibited more phagocytosis. However, internalization of PA+Sia is associated with a reduction in respiratory burst and increase in anti-inflammatory cytokines secretion which is reversed upon desialylation of the bacteria. Phagocytosis of PA+Sia is also associated with reduced intracellular calcium ion concentrations and altered calcium-dependent signaling which negatively affects phagosome maturation. Consequently, although more PA+Sia was localized in early phagosomes (Rab5 compartment), only fewer bacteria reach into the late phagosomal compartment (Rab7). Possibly, this leads to reduced phagosome lysosome fusion where reduced numbers of PA+Sia are trafficked into lysosomes, compared to PA−Sia. Thus, internalized PA+Sia remain viable and replicates intracellularly in macrophages. We have also demonstrated that such siglec-E-sialic acid interaction recruited SHP-1/SHP-2 phosphatases which modulate MAPK and NF-κB signaling pathways. Disrupting sialic acid-siglec-E interaction by silencing siglec-E in macrophages results in improved bactericidal response against PA+Sia characterized by robust respiratory burst, enhanced intracellular calcium levels and nuclear translocation of p65 component of NF-κB complex leading to increased pro-inflammatory cytokine secretion. Taken together, we have identified that sialic acid-siglec-E interactions is another pathway utilized by PA in order to suppress macrophage antimicrobial responses and inhibit phagosome maturation, thereby persisting as an intracellular pathogen in macrophages.
Collapse
Affiliation(s)
- Kaustuv Mukherjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Biswajit Khatua
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
9
|
Pradhan G, Raj Abraham P, Shrivastava R, Mukhopadhyay S. Calcium Signaling Commands Phagosome Maturation Process. Int Rev Immunol 2020; 38:57-69. [PMID: 31117900 DOI: 10.1080/08830185.2019.1592169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phagosome-lysosome (P-L) fusion is one of the central immune-effector responses of host. It is known that phagosome maturation process is associated with numerous signaling cascades and among these, important role of calcium (Ca2+) signaling has been realized recently. Ca2+ plays key roles in actin rearrangement, activation of NADPH oxidase and protein kinase C (PKC). Involvement of Ca2+ in these cellular processes directs phagosomal maturation process. Some of the intracellular pathogens have acquired the strategies to modulate Ca2+ associated pathways to block P-L fusion process. In this review we have described the mechanism of Ca2+ signals that influence P-L fusion by controlling ROS, actin and PKC signaling cascades. We have also discussed the strategies implemented by the intracellular pathogens to manipulate Ca2+ signaling to consequently subvert P-L fusion. A detail study of factors associated in manipulating Ca2+ signaling may provide new insights for the development of therapeutic tools for more effective treatment options against infectious diseases.
Collapse
Affiliation(s)
- Gourango Pradhan
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India.,b Graduate Studies , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Philip Raj Abraham
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India
| | - Rohini Shrivastava
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India.,b Graduate Studies , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Sangita Mukhopadhyay
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India
| |
Collapse
|
10
|
Westman J, Grinstein S, Maxson ME. Revisiting the role of calcium in phagosome formation and maturation. J Leukoc Biol 2019; 106:837-851. [DOI: 10.1002/jlb.mr1118-444r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Johannes Westman
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| | - Sergio Grinstein
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
- Department of BiochemistryUniversity of Toronto Toronto Ontario Canada
- Keenan Research Centre of the Li Ka Shing Knowledge InstituteSt. Michael's Hospital Toronto Ontario Canada
| | - Michelle E. Maxson
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| |
Collapse
|
11
|
He H, Arsenault RJ, Genovese KJ, Swaggerty CL, Johnson C, Nisbet DJ, Kogut MH. Inhibition of calmodulin increases intracellular survival of Salmonella in chicken macrophage cells. Vet Microbiol 2019; 232:156-161. [PMID: 30967327 DOI: 10.1016/j.vetmic.2019.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 01/05/2023]
Abstract
Calcium (Ca2+) is a pivotal intracellular second messenger and calmodulin (CaM) acts as a multifunctional Ca2+-binding protein that regulates downstream Ca2+ dependent signaling. Together they play an important role in regulating various cellular functions, including gene expression, maturation of phagolysosome, apoptosis, and immune response. Intracellular Ca2+ has been shown to play a critical role in Toll-like receptor-mediated immune response to microbial agonists in the HD11 chicken macrophage cell line. The role of that the Ca2+/CaM pathway plays in the intracellular survival of Salmonella in chicken macrophages has not been reported. In this study, kinome peptide array analysis indicated that the Ca2+/CaM pathway was significantly activated when chicken macrophage HD11 cells were infected with S. Enteritidis or S. Heidelberg. Further study demonstrated that treating cells with a pharmaceutical CaM inhibitor W-7, which disrupts the formation of Ca2+/CaM, significantly inhibited macrophages to produce nitric oxide and weaken the control of intracellular Salmonella replication. These results strongly indicate that CaM plays an important role in the innate immune response of chicken macrophages and that the Ca2+/CaM mediated signaling pathway is critically involved in the host cell response to Salmonella infection.
Collapse
Affiliation(s)
- Haiqi He
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX 77845, United States.
| | - Ryan J Arsenault
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, United States
| | - Kenneth J Genovese
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX 77845, United States
| | - Christina L Swaggerty
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX 77845, United States
| | - Casey Johnson
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, United States
| | - David J Nisbet
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX 77845, United States
| | - Michael H Kogut
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX 77845, United States
| |
Collapse
|
12
|
Dingjan I, Linders PTA, Verboogen DRJ, Revelo NH, Ter Beest M, van den Bogaart G. Endosomal and Phagosomal SNAREs. Physiol Rev 2018; 98:1465-1492. [PMID: 29790818 DOI: 10.1152/physrev.00037.2017] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein family is of vital importance for organelle communication. The complexing of cognate SNARE members present in both the donor and target organellar membranes drives the membrane fusion required for intracellular transport. In the endocytic route, SNARE proteins mediate trafficking between endosomes and phagosomes with other endosomes, lysosomes, the Golgi apparatus, the plasma membrane, and the endoplasmic reticulum. The goal of this review is to provide an overview of the SNAREs involved in endosomal and phagosomal trafficking. Of the 38 SNAREs present in humans, 30 have been identified at endosomes and/or phagosomes. Many of these SNAREs are targeted by viruses and intracellular pathogens, which thereby reroute intracellular transport for gaining access to nutrients, preventing their degradation, and avoiding their detection by the immune system. A fascinating picture is emerging of a complex transport network with multiple SNAREs being involved in consecutive trafficking routes.
Collapse
Affiliation(s)
- Ilse Dingjan
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Peter T A Linders
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Danielle R J Verboogen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
13
|
Kim ST, Lee YJ, Tasaki T, Mun SR, Hwang J, Kang MJ, Ganipisetti S, Yi EC, Kim BY, Kwon YT. The N-recognin UBR4 of the N-end rule pathway is targeted to and required for the biogenesis of the early endosome. J Cell Sci 2018; 131:jcs.217646. [PMID: 30111582 DOI: 10.1242/jcs.217646] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/30/2018] [Indexed: 12/26/2022] Open
Abstract
The N-end rule pathway is a proteolytic system in which single N-terminal residues of proteins act as N-degrons. These degrons are recognized by N-recognins, facilitating substrate degradation via the ubiquitin (Ub) proteasome system (UPS) or autophagy. We have previously identified a set of N-recognins [UBR1, UBR2, UBR4 (also known as p600) and UBR5 (also known as EDD)] that bind N-degrons through their UBR boxes to promote proteolysis by the proteasome. Here, we show that the 570 kDa N-recognin UBR4 is associated with maturing endosomes through an interaction with Ca2+-bound calmodulin. The endosomal recruitment of UBR4 is essential for the biogenesis of early endosomes (EEs) and endosome-related processes, such as the trafficking of endocytosed protein cargos and degradation of extracellular cargos by endosomal hydrolases. In mouse embryos, UBR4 marks and plays a role in the endosome-lysosome pathway that mediates the heterophagic proteolysis of endocytosed maternal proteins into amino acids. By screening 9591 drugs through the DrugBank database, we identify picolinic acid as a putative ligand for UBR4 that inhibits the biogenesis of EEs. Our results suggest that UBR4 is an essential modulator in the endosome-lysosome system.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sung Tae Kim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States
| | - Yoon Jee Lee
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Takafumi Tasaki
- Division of Protein Regulation Research, Medical Research Institute, Kanazawa Medical University, Ishikawa, 920-0293, Japan.,Department of Medical Zoology, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Su Ran Mun
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Joonsung Hwang
- World Class Institute, Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, 28116, Republic of Korea
| | - Min Jueng Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, 03080, Republic of Korea
| | - Srinivasrao Ganipisetti
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, 03080, Republic of Korea
| | - Bo Yeon Kim
- World Class Institute, Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, 28116, Republic of Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea .,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
14
|
The anti-tumor drug 2-hydroxyoleic acid (Minerval) stimulates signaling and retrograde transport. Oncotarget 2018; 7:86871-86888. [PMID: 27894086 PMCID: PMC5349960 DOI: 10.18632/oncotarget.13508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/17/2016] [Indexed: 11/25/2022] Open
Abstract
2-hydroxyoleic acid (OHOA, Minerval®) is an example of a substance used for membrane lipid therapy, where the cellular membranes rather than specific proteins constitute the therapeutical target. OHOA is thought to mediate its anti-tumor effect by affecting the biophysical properties of membranes, which leads to altered recruitment and activation of amphitropic proteins, altered cellular signaling, and eventual cell death. Little is known about the initial signaling events upon treatment with OHOA, and whether the altered membrane properties would have any impact on the dynamic intracellular transport system. In the present study we demonstrate that treatment with OHOA led to a rapid release of intracellular calcium and activation of multiple signaling pathways in HeLa cells, including the PI3K-AKT1-MTOR pathway and several MAP kinases, in a process independent of the EGFR. By lipidomics we confirmed that OHOA was incorporated into several lipid classes. Concomitantly, OHOA potently increased retrograde transport of the plant toxin ricin from endosomes to the Golgi and further to the endoplasmic reticulum. The OHOA-stimulated ricin transport seemed to require several amphitropic proteins, including Src, phospholipase C, protein kinase C, and also Ca2+/calmodulin. Interestingly, OHOA induced a slight increase in endosomal localization of the retromer component VPS35. Thus, our data show that addition of a lipid known to alter membrane properties not only affects signaling, but also intracellular transport.
Collapse
|
15
|
Sun X, Yang Y, Zhong XZ, Cao Q, Zhu XH, Zhu X, Dong XP. A negative feedback regulation of MTORC1 activity by the lysosomal Ca 2+ channel MCOLN1 (mucolipin 1) using a CALM (calmodulin)-dependent mechanism. Autophagy 2018; 14:38-52. [PMID: 29460684 DOI: 10.1080/15548627.2017.1389822] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway that is required for cellular homeostasis, growth and survival. The lysosome plays an essential role in autophagy regulation. For example, the activity of MTORC1, a master regulator of autophagy, is regulated by nutrients within the lysosome. Starvation inhibits MTORC1 causing autophagy induction. Given that MTORC1 is critical for protein synthesis and cellular homeostasis, a feedback regulatory mechanism must exist to restore MTORC1 during starvation. However, the molecular mechanism underlying this feedback regulation is unclear. In this study, we report that starvation activates the lysosomal Ca2+ release channel MCOLN1 (mucolipin 1) by relieving MTORC1's inhibition of the channel. Activated MCOLN1 in turn facilitates MTORC1 activity that requires CALM (calmodulin). Moreover, both MCOLN1 and CALM are necessary for MTORC1 reactivation during prolonged starvation. Our data suggest that lysosomal Ca2+ signaling is an essential component of the canonical MTORC1-dependent autophagy pathway and MCOLN1 provides a negative feedback regulation of MTORC1 to prevent excessive loss of MTORC1 function during starvation. The feedback regulation may be important for maintaining cellular homeostasis during starvation, as well as many other stressful or disease conditions.
Collapse
Affiliation(s)
- Xue Sun
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada.,d Key Laboratory of Molecular Epigenetics of Ministry of Education , Institute of Cytology and Genetics, Northeast Normal University , Changchun , Jilin , China
| | - Yiming Yang
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Xi Zoë Zhong
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Qi Cao
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Xin-Hong Zhu
- b Institute of Mental Health, Southern Medical University , Guangzhou , China.,c Key Laboratory of Psychiatric Disorders of Guangdong Province , Guangzhou , China
| | - Xiaojuan Zhu
- d Key Laboratory of Molecular Epigenetics of Ministry of Education , Institute of Cytology and Genetics, Northeast Normal University , Changchun , Jilin , China
| | - Xian-Ping Dong
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| |
Collapse
|
16
|
Hauswirth AG, Ford KJ, Wang T, Fetter RD, Tong A, Davis GW. A postsynaptic PI3K-cII dependent signaling controller for presynaptic homeostatic plasticity. eLife 2018; 7:31535. [PMID: 29303480 PMCID: PMC5773188 DOI: 10.7554/elife.31535] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/04/2018] [Indexed: 01/29/2023] Open
Abstract
Presynaptic homeostatic plasticity stabilizes information transfer at synaptic connections in organisms ranging from insect to human. By analogy with principles of engineering and control theory, the molecular implementation of PHP is thought to require postsynaptic signaling modules that encode homeostatic sensors, a set point, and a controller that regulates transsynaptic negative feedback. The molecular basis for these postsynaptic, homeostatic signaling elements remains unknown. Here, an electrophysiology-based screen of the Drosophila kinome and phosphatome defines a postsynaptic signaling platform that includes a required function for PI3K-cII, PI3K-cIII and the small GTPase Rab11 during the rapid and sustained expression of PHP. We present evidence that PI3K-cII localizes to Golgi-derived, clathrin-positive vesicles and is necessary to generate an endosomal pool of PI(3)P that recruits Rab11 to recycling endosomal membranes. A morphologically distinct subdivision of this platform concentrates postsynaptically where we propose it functions as a homeostatic controller for retrograde, trans-synaptic signaling.
Collapse
Affiliation(s)
- Anna G Hauswirth
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Kevin J Ford
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Tingting Wang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Amy Tong
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
17
|
Gabriel M, Moya-Díaz J, Gallo LI, Marengo FD, Estrada LC. Single particle tracking of internalized metallic nanoparticles reveals heterogeneous directed motion after clathrin dependent endocytosis in mouse chromaffin cells. Methods Appl Fluoresc 2017; 6:014003. [PMID: 28901956 DOI: 10.1088/2050-6120/aa8c64] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Most accepted single particle tracking methods are able to obtain high-resolution trajectories for relatively short periods of time. In this work we apply a straightforward combination of single-particle tracking microscopy and metallic nanoparticles internalization on mouse chromaffin cells to unveil the intracellular trafficking mechanism of metallic-nanoparticle-loaded vesicles (MNP-V) complexes after clathrin dependent endocytosis. We found that directed transport is the major route of MNP-Vs intracellular trafficking after stimulation (92.6% of the trajectories measured). We then studied the MNP-V speed at each point along the trajectory, and found that the application of a second depolarization stimulus during the tracking provokes an increase in the percentage of low-speed trajectory points in parallel with a decrease in the number of high-speed trajectory points. This result suggests that stimulation may facilitate the compartmentalization of internalized MNPs in a more restricted location such as was already demonstrated in neuronal and neuroendocrine cells (Bronfman et al 2003 J. Neurosci. 23 3209-20). Although further experiments will be required to address the mechanisms underlying this transport dynamics, our studies provide quantitative evidence of the heterogeneous behavior of vesicles mobility after endocytosis in chromaffin cells highlighting the potential of MNPs as alternative labels in optical microscopy to provide new insights into the vesicles dynamics in a wide variety of cellular environments.
Collapse
Affiliation(s)
- Manuela Gabriel
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Ciudad Universitaria, 1428 Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
18
|
Konopka-Postupolska D, Clark G. Annexins as Overlooked Regulators of Membrane Trafficking in Plant Cells. Int J Mol Sci 2017; 18:E863. [PMID: 28422051 PMCID: PMC5412444 DOI: 10.3390/ijms18040863] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022] Open
Abstract
Annexins are an evolutionary conserved superfamily of proteins able to bind membrane phospholipids in a calcium-dependent manner. Their physiological roles are still being intensively examined and it seems that, despite their general structural similarity, individual proteins are specialized toward specific functions. However, due to their general ability to coordinate membranes in a calcium-sensitive fashion they are thought to participate in membrane flow. In this review, we present a summary of the current understanding of cellular transport in plant cells and consider the possible roles of annexins in different stages of vesicular transport.
Collapse
Affiliation(s)
- Dorota Konopka-Postupolska
- Plant Biochemistry Department, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw 02-106, Poland.
| | - Greg Clark
- Molecular, Cell, and Developmental Biology, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
19
|
Cao Q, Yang Y, Zhong XZ, Dong XP. The lysosomal Ca 2+ release channel TRPML1 regulates lysosome size by activating calmodulin. J Biol Chem 2017; 292:8424-8435. [PMID: 28360104 DOI: 10.1074/jbc.m116.772160] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/29/2017] [Indexed: 01/01/2023] Open
Abstract
Intracellular lysosomal membrane trafficking, including fusion and fission, is crucial for cellular homeostasis and normal cell function. Both fusion and fission of lysosomal membrane are accompanied by lysosomal Ca2+ release. We recently have demonstrated that the lysosomal Ca2+ release channel P2X4 regulates lysosome fusion through a calmodulin (CaM)-dependent mechanism. However, the molecular mechanism underlying lysosome fission remains uncertain. In this study, we report that enlarged lysosomes/vacuoles induced by either vacuolin-1 or P2X4 activation are suppressed by up-regulating the lysosomal Ca2+ release channel transient receptor potential mucolipin 1 (TRPML1) but not the lysosomal Na+ release channel two-pore channel 2 (TPC2). Activation of TRPML1 facilitated the recovery of enlarged lysosomes/vacuoles. Moreover, the effects of TRPML1 on lysosome/vacuole size regulation were eliminated by Ca2+ chelation, suggesting a requirement for TRPML1-mediated Ca2+ release. We further demonstrate that the prototypical Ca2+ sensor CaM is required for the regulation of lysosome/vacuole size by TRPML1, suggesting that TRPML1 may promote lysosome fission by activating CaM. Given that lysosome fission is implicated in both lysosome biogenesis and reformation, our findings suggest that TRPML1 may function as a key lysosomal Ca2+ channel controlling both lysosome biogenesis and reformation.
Collapse
Affiliation(s)
- Qi Cao
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Yiming Yang
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Xi Zoë Zhong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
20
|
Woo SS, James DJ, Martin TFJ. Munc13-4 functions as a Ca 2+ sensor for homotypic secretory granule fusion to generate endosomal exocytic vacuoles. Mol Biol Cell 2017; 28:792-808. [PMID: 28100639 PMCID: PMC5349786 DOI: 10.1091/mbc.e16-08-0617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/06/2017] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
Munc13-4 is a Ca2+-dependent SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)- and phospholipid-binding protein that localizes to and primes secretory granules (SGs) for Ca2+-evoked secretion in various secretory cells. Studies in mast cell-like RBL-2H3 cells provide direct evidence that Munc13-4 with its two Ca2+-binding C2 domains functions as a Ca2+ sensor for SG exocytosis. Unexpectedly, Ca2+ stimulation also generated large (>2.4 μm in diameter) Munc13-4+/Rab7+/Rab11+ endosomal vacuoles. Vacuole generation involved the homotypic fusion of Munc13-4+/Rab7+ SGs, followed by a merge with Rab11+ endosomes, and depended on Ca2+ binding to Munc13-4. Munc13-4 promoted the Ca2+-stimulated fusion of VAMP8-containing liposomes with liposomes containing exocytic or endosomal Q-SNAREs and directly interacted with late endosomal SNARE complexes. Thus Munc13-4 is a tethering/priming factor and Ca2+ sensor for both heterotypic SG-plasma membrane and homotypic SG-SG fusion. Total internal reflection fluorescence microscopy imaging revealed that vacuoles were exocytic and mediated secretion of β-hexosaminidase and cytokines accompanied by Munc13-4 diffusion onto the plasma membrane. The results provide new molecular insights into the mechanism of multigranular compound exocytosis commonly observed in various secretory cells.
Collapse
Affiliation(s)
- Sang Su Woo
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Declan J James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Thomas F J Martin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
21
|
Enrich C, Rentero C, Grewal T. Annexin A6 in the liver: From the endocytic compartment to cellular physiology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:933-946. [PMID: 27984093 DOI: 10.1016/j.bbamcr.2016.10.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/15/2022]
Abstract
Annexin A6 (AnxA6) belongs to the conserved annexin family - a group of Ca2+-dependent membrane binding proteins. AnxA6 is the largest of all annexins and highly expressed in smooth muscle, hepatocytes, endothelial cells and cardiomyocytes. Upon activation, AnxA6 binds to negatively charged phospholipids in a wide range of intracellular localizations, in particular the plasma membrane, late endosomes/pre-lysosomes, but also synaptic vesicles and sarcolemma. In these cellular sites, AnxA6 is believed to contribute to the organization of membrane microdomains, such as cholesterol-rich lipid rafts and confer multiple regulatory functions, ranging from vesicle fusion, endocytosis and exocytosis to programmed cell death and muscle contraction. Growing evidence supports that Ca2+ and Ca2+-binding proteins control endocytosis and autophagy. Their regulatory role seems to operate at the level of the signalling pathways that initiate autophagy or at later stages, when autophagosomes fuse with endolysosomal compartments. The convergence of the autophagic and endocytic vesicles to lysosomes shares several features that depend on Ca2+ originating from lysosomes/late endosomes and seems to depend on proteins that are subsequently activated by this cation. However, the involvement of Ca2+ and its effector proteins in these autophagic and endocytic stages still remains poorly understood. Although AnxA6 makes up almost 0.25% of total protein in the liver, little is known about its function in hepatocytes. Within the endocytic route, we identified AnxA6 in endosomes and autophagosomes of hepatocytes. Hence, AnxA6 and possibly other annexins might represent new Ca2+ effectors that regulate converging steps of autophagy and endocytic trafficking in hepatocytes. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cellular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain.
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cellular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy A15, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
Rayl M, Truitt M, Held A, Sargeant J, Thorsen K, Hay JC. Penta-EF-Hand Protein Peflin Is a Negative Regulator of ER-To-Golgi Transport. PLoS One 2016; 11:e0157227. [PMID: 27276012 PMCID: PMC4898701 DOI: 10.1371/journal.pone.0157227] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/26/2016] [Indexed: 11/18/2022] Open
Abstract
Luminal calcium regulates vesicle transport early in the secretory pathway. In ER-to-Golgi transport, depletion of luminal calcium leads to significantly reduced transport and a buildup of budding and newly budded COPII vesicles and vesicle proteins. Effects of luminal calcium on transport may be mediated by cytoplasmic calcium sensors near ER exits sites (ERES). The penta-EF-hand (PEF) protein apoptosis-linked gene 2 (ALG-2) stabilizes sec31A at ER exit sites (ERES) and promotes the assembly of inner and outer shell COPII components. However, in vitro and intact cell approaches have not determined whether ALG-2 is a negative or positive regulator, or a regulator at all, under basal physiological conditions. ALG-2 interacts with another PEF protein, peflin, to form cytosolic heterodimers that dissociate in response to calcium. However, a biological function for peflin has not been demonstrated and whether peflin and the ALG-2/peflin interaction modulates transport has not been investigated. Using an intact, single cell, morphological assay for ER-to-Golgi transport in normal rat kidney (NRK) cells, we found that depletion of peflin using siRNA resulted in significantly faster transport of the membrane cargo VSV-G. Double depletion of peflin and ALG-2 blocked the increased transport resulting from peflin depletion, demonstrating a role for ALG-2 in the increased transport. Furthermore, peflin depletion caused increased targeting of ALG-2 to ERES and increased ALG-2/sec31A interactions, suggesting that peflin may normally inhibit transport by preventing ALG-2/sec31A interactions. This work identifies for the first time a clear steady state role for a PEF protein in ER-to-Golgi transport—peflin is a negative regulator of transport.
Collapse
Affiliation(s)
- Mariah Rayl
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
| | - Mishana Truitt
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
| | - Aaron Held
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
| | - John Sargeant
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
| | - Kevin Thorsen
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
| | - Jesse C. Hay
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, United States of America
- * E-mail:
| |
Collapse
|
23
|
Mukherjee I, Barlowe C. Overexpression of Sly41 suppresses COPII vesicle-tethering deficiencies by elevating intracellular calcium levels. Mol Biol Cell 2016; 27:1635-49. [PMID: 27030673 PMCID: PMC4865320 DOI: 10.1091/mbc.e15-10-0704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/22/2016] [Indexed: 11/29/2022] Open
Abstract
SLY41 is a multicopy suppressor of mutations in the essential Ypt1 GTPase. Overexpression of Sly41 elevates cytosolic Ca2+ concentration, which stimulates SNARE-dependent fusion of COPII vesicles with Golgi membranes and suppresses deficiencies in Ypt1-dependent vesicle tethering. Thus Ca2+ positively regulates vesicle fusion with Golgi membranes. SLY41 was identified as a multicopy suppressor of loss of Ypt1, a Rab GTPase essential for COPII vesicle tethering at the Golgi complex. SLY41 encodes a polytopic membrane protein with homology to a class of solute transporter proteins, but how overexpression suppresses vesicle-tethering deficiencies is not known. Here we show that Sly41 is efficiently packaged into COPII vesicles and actively cycles between the ER and Golgi compartments. SLY41 displays synthetic negative genetic interactions with PMR1, which encodes the major Golgi-localized Ca2+/Mn2+ transporter and suggests that Sly41 influences cellular Ca2+ and Mn2+ homeostasis. Experiments using the calcium probe aequorin to measure intracellular Ca2+ concentrations in live cells reveal that Sly41 overexpression significantly increases cytosolic calcium levels. Although specific substrates of the Sly41 transporter were not identified, our findings indicate that localized overexpression of Sly41 to the early secretory pathway elevates cytosolic calcium levels to suppress vesicle-tethering mutants. In vitro SNARE cross-linking assays were used to directly monitor the influence of Ca2+ on tethering and fusion of COPII vesicles with Golgi membranes. Strikingly, calcium at suppressive concentrations stimulated SNARE-dependent membrane fusion when vesicle-tethering activity was reduced. These results show that calcium positively regulates the SNARE-dependent fusion stage of ER–Golgi transport.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Charles Barlowe
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
24
|
Mittal R, Chan B, Grati M, Mittal J, Patel K, Debs LH, Patel AP, Yan D, Chapagain P, Liu XZ. Molecular Structure and Regulation of P2X Receptors With a Special Emphasis on the Role of P2X2 in the Auditory System. J Cell Physiol 2015; 231:1656-70. [PMID: 26627116 DOI: 10.1002/jcp.25274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/01/2015] [Indexed: 12/23/2022]
Abstract
The P2X purinergic receptors are cation-selective channels gated by extracellular adenosine 5'-triphosphate (ATP). These purinergic receptors are found in virtually all mammalian cell types and facilitate a number of important physiological processes. Within the past few years, the characterization of crystal structures of the zebrafish P2X4 receptor in its closed and open states has provided critical insights into the mechanisms of ligand binding and channel activation. Understanding of this gating mechanism has facilitated to design and interpret new modeling and structure-function experiments to better elucidate how different agonists and antagonists can affect the receptor with differing levels of potency. This review summarizes the current knowledge on the structure, activation, allosteric modulators, function, and location of the different P2X receptors. Moreover, an emphasis on the P2X2 receptors has been placed in respect to its role in the auditory system. In particular, the discovery of three missense mutations in P2X2 receptors could become important areas of study in the field of gene therapy to treat progressive and noise-induced hearing loss. J. Cell. Physiol. 231: 1656-1670, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Brandon Chan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Kunal Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Luca H Debs
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Amit P Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, Florida.,Biomolecular Science Institute, Florida International University, Miami, Florida
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biochemistry, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
25
|
Cao Q, Zhong XZ, Zou Y, Murrell-Lagnado R, Zhu MX, Dong XP. Calcium release through P2X4 activates calmodulin to promote endolysosomal membrane fusion. J Cell Biol 2015; 209:879-94. [PMID: 26101220 PMCID: PMC4477861 DOI: 10.1083/jcb.201409071] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
P2X4 and calmodulin form a signaling complex in late endosomes and lysosomes that promotes fusion and vacuolation in a Ca2+-dependent fashion. Intra-endolysosomal Ca2+ release is required for endolysosomal membrane fusion with intracellular organelles. However, the molecular mechanisms for intra-endolysosomal Ca2+ release and the downstream Ca2+ targets involved in the fusion remain elusive. Previously, we demonstrated that endolysosomal P2X4 forms channels activated by luminal adenosine triphosphate in a pH-dependent manner. In this paper, we show that overexpression of P2X4, as well as increasing endolysosomal P2X4 activity by alkalinization of endolysosome lumen, promoted vacuole enlargement in cells and endolysosome fusion in a cell-free assay. These effects were prevented by inhibiting P2X4, expressing a dominant-negative P2X4 mutant, and disrupting the P2X4 gene. We further show that P2X4 and calmodulin (CaM) form a complex at endolysosomal membrane where P2X4 activation recruits CaM to promote fusion and vacuolation in a Ca2+-dependent fashion. Moreover, P2X4 activation-triggered fusion and vacuolation were suppressed by inhibiting CaM. Our data thus suggest a new molecular mechanism for endolysosomal membrane fusion involving P2X4-mediated endolysosomal Ca2+ release and subsequent CaM activation.
Collapse
Affiliation(s)
- Qi Cao
- Department of Physiology and Biophysics, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Xi Zoë Zhong
- Department of Physiology and Biophysics, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Yuanjie Zou
- Department of Physiology and Biophysics, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Ruth Murrell-Lagnado
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| |
Collapse
|
26
|
Helm JR, Bentley M, Thorsen KD, Wang T, Foltz L, Oorschot V, Klumperman J, Hay JC. Apoptosis-linked gene-2 (ALG-2)/Sec31 interactions regulate endoplasmic reticulum (ER)-to-Golgi transport: a potential effector pathway for luminal calcium. J Biol Chem 2014; 289:23609-28. [PMID: 25006245 DOI: 10.1074/jbc.m114.561829] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Luminal calcium released from secretory organelles has been suggested to play a regulatory role in vesicle transport at several steps in the secretory pathway; however, its functional roles and effector pathways have not been elucidated. Here we demonstrate for the first time that specific luminal calcium depletion leads to a significant decrease in endoplasmic reticulum (ER)-to-Golgi transport rates in intact cells. Ultrastructural analysis revealed that luminal calcium depletion is accompanied by increased accumulation of intermediate compartment proteins in COPII buds and clusters of unfused COPII vesicles at ER exit sites. Furthermore, we present several lines of evidence suggesting that luminal calcium affected transport at least in part through calcium-dependent interactions between apoptosis-linked gene-2 (ALG-2) and the Sec31A proline-rich region: 1) targeted disruption of ALG-2/Sec31A interactions caused severe defects in ER-to-Golgi transport in intact cells; 2) effects of luminal calcium and ALG-2/Sec31A interactions on transport mutually required each other; and 3) Sec31A function in transport required luminal calcium. Morphological phenotypes of disrupted ALG-2/Sec31A interactions were characterized. We found that ALG-2/Sec31A interactions were not required for the localization of Sec31A to ER exit sites per se but appeared to acutely regulate the stability and trafficking of the cargo receptor p24 and the distribution of the vesicle tether protein p115. These results represent the first outline of a mechanism that connects luminal calcium to specific protein interactions regulating vesicle trafficking machinery.
Collapse
Affiliation(s)
- Jared R Helm
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| | - Marvin Bentley
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| | - Kevin D Thorsen
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| | - Ting Wang
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| | - Lauren Foltz
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| | - Viola Oorschot
- the Cell Microscopy Center, Department of Cell Biology, University Medical Center Utrecht, AZU Room H02.313, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Judith Klumperman
- the Cell Microscopy Center, Department of Cell Biology, University Medical Center Utrecht, AZU Room H02.313, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jesse C Hay
- From the Division of Biological Sciences and Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana 59812-4824 and
| |
Collapse
|
27
|
Pandey K, Dhoke RR, Rathore YS, Nath SK, Verma N, Bawa S, Ashish. Low pH Overrides the Need of Calcium Ions for the Shape–Function Relationship of Calmodulin: Resolving Prevailing Debates. J Phys Chem B 2014; 118:5059-74. [DOI: 10.1021/jp501641r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kalpana Pandey
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Reema R. Dhoke
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | | | - Samir K. Nath
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Neha Verma
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Simranjot Bawa
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Ashish
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| |
Collapse
|
28
|
van der Kant R, Neefjes J. Small regulators, major consequences - Ca²⁺ and cholesterol at the endosome-ER interface. J Cell Sci 2014; 127:929-38. [PMID: 24554437 DOI: 10.1242/jcs.137539] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The ER is the largest cellular compartment and a major storage site for lipids and ions. In recent years, much attention has focused on contacts between the ER and other organelles, and one particularly intimate relationship is that between the ER and the endosomal system. ER-endosome contacts intensify when endosomes mature, and the ER participates in endosomal processes, such as the termination of surface receptor signaling, multi-vesicular body formation, and transport and fusion events. Cholesterol and Ca(2+) are transferred between the ER and endosomes, possibly acting as messengers for ER-endosome crosstalk. Here, we summarize different types of ER-endosomal communication and discuss membrane contact sites that might facilitate this crosstalk. We review the protein pairs that interact at the ER-endosome interface and find that many of these have a role in cholesterol exchange. We also summarize Ca(2+) exchange between the ER and endosomes, and hypothesize that ER-endosome contacts integrate several cellular functions to guide endosomal maturation. We post the hypothesis that failure in ER-endosome contacts is an unrecognized but important contributor to diseases, such as Niemann-Pick type C disease, Alzheimer's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Rik van der Kant
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
29
|
Ghislat G, Knecht E. Ca²⁺-sensor proteins in the autophagic and endocytic traffic. Curr Protein Pept Sci 2014; 14:97-110. [PMID: 23305313 PMCID: PMC3664516 DOI: 10.2174/13892037112139990033] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/19/2012] [Accepted: 11/28/2012] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis are two evolutionarily conserved catabolic processes that comprise vesicle trafficking events for the clearance of the sequestered intracellular and extracellular cargo. Both start differently but end in the same compartment, the lysosome. Mounting evidences from the last years have established the involvement of proteins sensitive to intracellular Ca2+ in the control of the early autophagic steps and in the traffic of autophagic, endocytic and lysosomal vesicles. However, this knowledge is based on dispersed outcomes that do not set up a consensus model of the Ca2+-dependent control of autophagy and endocytosis. Here, we will provide a critical synopsis of insights from the last decade on the involvement of Ca2+-sensor proteins in the activation of autophagy and in fusion events of endocytic vesicles, autophagosomes and lysosomes.
Collapse
Affiliation(s)
- Ghita Ghislat
- Laboratorio de Biología Celular, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain and CIBERER, Valencia, Spain
| | | |
Collapse
|
30
|
Parkinson K, Baines AE, Keller T, Gruenheit N, Bragg L, North RA, Thompson CR. Calcium-dependent regulation of Rab activation and vesicle fusion by an intracellular P2X ion channel. Nat Cell Biol 2014; 16:87-98. [PMID: 24335649 PMCID: PMC3942655 DOI: 10.1038/ncb2887] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/30/2013] [Indexed: 11/24/2022]
Abstract
Rab GTPases play key roles in the delivery, docking and fusion of intracellular vesicles. However, the mechanism by which spatial and temporal regulation of Rab GTPase activity is controlled is poorly understood. Here we describe a mechanism by which localized calcium release through a vesicular ion channel controls Rab GTPase activity. We show that activation of P2XA, an intracellular ion channel localized to the Dictyostelium discoideum contractile vacuole system, results in calcium efflux required for downregulation of Rab11a activity and efficient vacuole fusion. Vacuole fusion and Rab11a downregulation require the activity of CnrF, an EF-hand-containing Rab GAP found in a complex with Rab11a and P2XA. CnrF Rab GAP activity for Rab11a is enhanced by the presence of calcium and the EF-hand domain. These findings suggest that P2XA activation results in vacuolar calcium release, which triggers activation of CnrF Rab GAP activity and subsequent downregulation of Rab11a to allow vacuole fusion.
Collapse
Affiliation(s)
- Katie Parkinson
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Abigail E. Baines
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Thomas Keller
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Nicole Gruenheit
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Laricia Bragg
- Faculty of Medical and Human Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - R. Alan North
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
- Faculty of Medical and Human Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Christopher R.L. Thompson
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| |
Collapse
|
31
|
Ando K, Kudo Y, Aoyagi K, Ishikawa R, Igarashi M, Takahashi M. Calmodulin-dependent regulation of neurotransmitter release differs in subsets of neuronal cells. Brain Res 2013; 1535:1-13. [DOI: 10.1016/j.brainres.2013.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/25/2013] [Accepted: 08/08/2013] [Indexed: 02/05/2023]
|
32
|
Troulinaki K, Tavernarakis N. Necrotic cell death and neurodegeneration: The involvement of endocytosis and intracellular trafficking. WORM 2013; 1:176-81. [PMID: 24058844 PMCID: PMC3670410 DOI: 10.4161/worm.20457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 04/20/2012] [Indexed: 11/26/2022]
Abstract
Necrosis, one of the two main types of cell death, contributes critically in many devastating pathological conditions in human, including stroke, ischemia, trauma and neurodegenerative diseases. However, unlike apoptosis, the molecular mechanisms underlying necrotic cell death and neurodegeneration are poorly understood. Caenorhabditis elegans offers a powerful platform for a thorough and systematic dissection of the molecular basis of necrotic cell death. Similarly to humans, neuronal necrosis can be induced by several well-characterized genetic lesions and by adverse environmental conditions in the nematode. The availability of precisely-defined C. elegans neurodegeneration models provides a unique opportunity for comprehensive delineation of the cellular and molecular mechanisms mediating necrotic cell death. Through genetic dissection of such models, we recently uncovered an unexpected requirement for specific proteins involved in endocytosis and intracellular trafficking, in the execution of necrosis. Moreover, initiation of necrotic cell death is accompanied by a sharp increase in the formation of early and recycling endosomes, which subsequently disintegrate during the final stage of cell death. These findings implicate endocytic and intracellular trafficking processes in the cellular destruction during necrosis. Indeed, endocytosis synergizes with two other essential cellular processes, autophagy and lysosomal proteolysis to facilitate necrotic neurodegeneration. In this commentary, we consider the contribution of endocytosis and intracellular trafficking to cell injury and discuss the crosstalk between these processes and other molecular mechanisms that mediate necrosis.
Collapse
Affiliation(s)
- Kostoula Troulinaki
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology; Heraklion, Crete Greece
| | | |
Collapse
|
33
|
Targeting multidrug-resistant tuberculosis (MDR-TB) by therapeutic vaccines. Med Microbiol Immunol 2012; 202:95-104. [DOI: 10.1007/s00430-012-0278-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 10/25/2012] [Indexed: 10/27/2022]
|
34
|
Fairn GD, Grinstein S. How nascent phagosomes mature to become phagolysosomes. Trends Immunol 2012; 33:397-405. [DOI: 10.1016/j.it.2012.03.003] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 03/15/2012] [Accepted: 03/24/2012] [Indexed: 01/18/2023]
|
35
|
Dennis AT, Nassal D, Deschenes I, Thomas D, Ficker E. Antidepressant-induced ubiquitination and degradation of the cardiac potassium channel hERG. J Biol Chem 2011; 286:34413-25. [PMID: 21832094 PMCID: PMC3190784 DOI: 10.1074/jbc.m111.254367] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 07/30/2011] [Indexed: 11/06/2022] Open
Abstract
The most common cause for adverse cardiac events by antidepressants is acquired long QT syndrome (acLQTS), which produces electrocardiographic abnormalities that have been associated with syncope, torsade de pointes arrhythmias, and sudden cardiac death. acLQTS is often caused by direct block of the cardiac potassium current I(Kr)/hERG, which is crucial for terminal repolarization in human heart. Importantly, desipramine belongs to a group of tricyclic antidepressant compounds that can simultaneously block hERG and inhibit its surface expression. Although up to 40% of all hERG blockers exert combined hERG block and trafficking inhibition, few of these compounds have been fully characterized at the cellular level. Here, we have studied in detail how desipramine inhibits hERG surface expression. We find a previously unrecognized combination of two entirely different mechanisms; desipramine increases hERG endocytosis and degradation as a consequence of drug-induced channel ubiquitination and simultaneously inhibits hERG forward trafficking from the endoplasmic reticulum. This unique combination of cellular effects in conjunction with acute channel block may explain why tricyclic antidepressants as a compound class are notorious for their association with arrhythmias and sudden cardiac death. Taken together, we describe the first example of drug-induced channel ubiquitination and degradation. Our data are directly relevant to the cardiac safety of not only tricyclic antidepressants but also other therapeutic compounds that exert multiple effects on hERG, as hERG trafficking and degradation phenotypes may go undetected in most preclinical safety assays designed to screen for acLQTS.
Collapse
Affiliation(s)
- Adrienne T. Dennis
- From the Rammelkamp Center for Education and Research, MetroHealth Campus, Case Western Reserve University, Cleveland Ohio 44109
| | - Drew Nassal
- From the Rammelkamp Center for Education and Research, MetroHealth Campus, Case Western Reserve University, Cleveland Ohio 44109
- the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland Ohio 44106, and
| | - Isabelle Deschenes
- From the Rammelkamp Center for Education and Research, MetroHealth Campus, Case Western Reserve University, Cleveland Ohio 44109
- the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland Ohio 44106, and
| | - Dierk Thomas
- the Department of Cardiology, Medical University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Eckhard Ficker
- From the Rammelkamp Center for Education and Research, MetroHealth Campus, Case Western Reserve University, Cleveland Ohio 44109
| |
Collapse
|
36
|
Vidal-Quadras M, Gelabert-Baldrich M, Soriano-Castell D, Lladó A, Rentero C, Calvo M, Pol A, Enrich C, Tebar F. Rac1 and Calmodulin Interactions Modulate Dynamics of ARF6-Dependent Endocytosis. Traffic 2011; 12:1879-96. [DOI: 10.1111/j.1600-0854.2011.01274.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Miller M, Dreisbach A, Otto A, Becher D, Bernhardt J, Hecker M, Peppelenbosch MP, van Dijl JM. Mapping of interactions between human macrophages and Staphylococcus aureus reveals an involvement of MAP kinase signaling in the host defense. J Proteome Res 2011; 10:4018-32. [PMID: 21736355 DOI: 10.1021/pr200224x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Staphylococcus aureus is a dangerous opportunistic human pathogen that causes serious invasive diseases when it reaches the bloodstream. Recent studies have shown that S. aureus is highly resistant to killing by professional phagocytes and that such cells even provide a favorable environment for intracellular survival of S. aureus. Importantly, the reciprocal interactions between phagocytes and S. aureus have remained largely elusive. Here we have employed kinase profiling to define the nature and time resolution of the human THP-1 macrophage response toward S. aureus and proteomics to identify the response of S. aureus toward macrophages. The results of these studies reveal major macrophage signaling pathways triggered by S. aureus and proteomic signatures of the responses of S. aureus to macrophages. We also identify human proteins bound to S. aureus that have potential roles in bacterial killing and internalization. Most noticeably, our observations challenge the classical concept that macrophage responses are mainly mediated through Toll-like receptor 2 and NF-κB signaling and highlight the important role of the stress-activated MAP kinase signaling in orchestrating the host defense.
Collapse
Affiliation(s)
- Malgorzata Miller
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen , Hanzeplein 1, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
38
|
de Keijzer S, Meddens MBM, Kilic D, Joosten B, Reinieren-Beeren I, Lidke DS, Cambi A. Interleukin-4 alters early phagosome phenotype by modulating class I PI3K dependent lipid remodeling and protein recruitment. PLoS One 2011; 6:e22328. [PMID: 21799824 PMCID: PMC3143135 DOI: 10.1371/journal.pone.0022328] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 06/24/2011] [Indexed: 11/19/2022] Open
Abstract
Phagocytosis is a complex process that involves membranelipid remodeling and the attraction and retention of key effector proteins. Phagosome phenotype depends on the type of receptor engaged and can be influenced by extracellular signals. Interleukin 4 (IL-4) is a cytokine that induces the alternative activation of macrophages (MΦs) upon prolonged exposure, triggering a different cell phenotype that has an altered phagocytic capacity. In contrast, the direct effects of IL-4 during phagocytosis remain unknown. Here, we investigate the impact of short-term IL-4 exposure (1 hour) during phagocytosis of IgG-opsonized yeast particles by MΦs. By time-lapse confocal microscopy of GFP-tagged lipid-sensing probes, we show that IL-4 increases the negative charge of the phagosomal membrane by prolonging the presence of the negatively charged second messenger PI(3,4,5)P3. Biochemical assays reveal an enhanced PI3K/Akt activity upon phagocytosis in the presence of IL-4. Blocking the specific class I PI3K after the onset of phagocytosis completely abrogates the IL-4-induced changes in lipid remodeling and concomitant membrane charge. Finally, we show that IL-4 direct signaling leads to a significantly prolonged retention profile of the signaling molecules Rac1 and Rab5 to the phagosomal membrane in a PI3K-dependent manner. This protracted early phagosome phenotype suggests an altered maturation, which is supported by the delayed phagosome acidification measured in the presence of IL-4. Our findings reveal that molecular differences in IL-4 levels, in the extracellular microenvironment, influence the coordination of lipid remodeling and protein recruitment, which determine phagosome phenotype and, eventually, fate. Endosomal and phagosomal membranes provide topological constraints to signaling molecules. Therefore, changes in the phagosome phenotype modulated by extracellular factors may represent an additional mechanism that regulates the outcome of phagocytosis and could have significant impact on the net biochemical output of a cell.
Collapse
Affiliation(s)
- Sandra de Keijzer
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Marjolein B. M. Meddens
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Dilek Kilic
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Ben Joosten
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Inge Reinieren-Beeren
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Diane S. Lidke
- Department of Pathology and Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Alessandra Cambi
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Mikhaylova M, Hradsky J, Kreutz MR. Between promiscuity and specificity: novel roles of EF-hand calcium sensors in neuronal Ca2+ signalling. J Neurochem 2011; 118:695-713. [PMID: 21722133 DOI: 10.1111/j.1471-4159.2011.07372.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In recent years, substantial progress has been made towards an understanding of the physiological function of EF-hand calcium sensor proteins of the Calmodulin (CaM) superfamily in neurons. This deeper appreciation is based on the identification of novel target interactions, structural studies and the discovery of novel signalling mechanisms in protein trafficking and synaptic plasticity, in which CaM-like sensor proteins appear to play a role. However, not all interactions are of plausible physiological relevance and in many cases it is not yet clear how the CaM signaling network relates to the proposed function of other EF-hand sensors. In this review, we will summarize these findings and address some of the open questions on the functional role of EF-hand calcium binding proteins in neurons.
Collapse
Affiliation(s)
- Marina Mikhaylova
- PG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | | | | |
Collapse
|
40
|
Calcium/calmodulin kinase II-dependent acetylcholine receptor cycling at the mammalian neuromuscular junction in vivo. J Neurosci 2010; 30:12455-65. [PMID: 20844140 DOI: 10.1523/jneurosci.3309-10.2010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
At the mammalian skeletal neuromuscular junction, cycling of nicotinic ACh receptors (nAChRs) is critical for the maintenance of a high postsynaptic receptor density. However, the mechanisms that regulate nAChRs recycling in living animals remain unknown. Using in vivo time-lapse imaging, fluorescence recovery after photobleaching, and biochemical pull down assays, we demonstrated that recycling of internalized nAChRs into fully functional and denervated synapses was promoted by both direct muscle stimulation and pharmacologically induced intracellular calcium elevations. Most of internalized nAChRs are recycled directly into synaptic sites. Chelating of intracellular calcium below resting level drastically decreased cycling of nAChRs. Furthermore we found that calcium-dependent AChR recycling is mediated by Ca(2+)/calmodulin-dependent kinase II (CaMKII). Inhibition of CaMKII selectively blocked recycling and caused intracellular accumulation of internalized nAChRs, whereas internalization of surface receptors remained unaffected. Electroporation of CaMKII-GFP isoforms into the sternomastoid muscle showed that muscle-specific CaMKIIβm isoform is highly expressed at the neuromuscular junction (NMJ) and precisely colocalized with nAChRs at crests of synaptic folds while the CaMKIIγ and δ isoforms are poorly expressed in synaptic sites. These results indicate that Ca(2+) along with CaMKII activity are critical for receptor recycling and may provide a mechanism by which the postsynaptic AChR density is maintained at the NMJ in vivo.
Collapse
|
41
|
Bentley M, Nycz DC, Joglekar A, Fertschai I, Malli R, Graier WF, Hay JC. Vesicular calcium regulates coat retention, fusogenicity, and size of pre-Golgi intermediates. Mol Biol Cell 2010; 21:1033-46. [PMID: 20089833 PMCID: PMC2836956 DOI: 10.1091/mbc.e09-10-0914] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This study establishes a role for luminal Ca2+ in ER/Golgi transport organelles and elucidates an effector mechanism involving the EF-hand protein ALG-2 and regulation of COPII coat retention. The significance and extent of Ca2+ regulation of the biosynthetic secretory pathway have been difficult to establish, and our knowledge of regulatory relationships integrating Ca2+ with vesicle coats and function is rudimentary. Here, we investigated potential roles and mechanisms of luminal Ca2+ in the early secretory pathway. Specific depletion of luminal Ca2+ in living normal rat kidney cells using cyclopiazonic acid (CPA) resulted in the extreme expansion of vesicular tubular cluster (VTC) elements. Consistent with this, a suppressive role for vesicle-associated Ca2+ in COPII vesicle homotypic fusion was demonstrated in vitro using Ca2+ chelators. The EF-hand–containing protein apoptosis-linked gene 2 (ALG-2), previously implicated in the stabilization of sec31 at endoplasmic reticulum exit sites, inhibited COPII vesicle fusion in a Ca2+-requiring manner, suggesting that ALG-2 may be a sensor for the effects of vesicular Ca2+ on homotypic fusion. Immunoisolation established that Ca2+ chelation inhibits and ALG-2 specifically favors residual retention of the COPII outer shell protein sec31 on pre-Golgi fusion intermediates. We conclude that vesicle-associated Ca2+, acting through ALG-2, favors the retention of residual coat molecules that seem to suppress membrane fusion. We propose that in cells, these Ca2+-dependent mechanisms temporally regulate COPII vesicle interactions, VTC biogenesis, cargo sorting, and VTC maturation.
Collapse
Affiliation(s)
- Marvin Bentley
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812-4824, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Nordenfelt P, Winberg ME, Lönnbro P, Rasmusson B, Tapper H. Different Requirements for Early and Late Phases of Azurophilic GranuleâPhagosome Fusion. Traffic 2009; 10:1881-93. [DOI: 10.1111/j.1600-0854.2009.00986.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
Zhao Z, Michaely P. The role of calcium in lipoprotein release by the low-density lipoprotein receptor. Biochemistry 2009; 48:7313-24. [PMID: 19583244 DOI: 10.1021/bi900214u] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The LDL receptor (LDLR) mediates efficient endocytosis of VLDL, VLDL remnants, and LDL. As part of the uptake process, the LDLR releases lipoproteins in endosomes. Released lipoproteins are subsequently trafficked to lysosomes for degradation, while the LDLR recycles back to the cell surface for further rounds of uptake. Endosomes have at least two features that can promote lipoprotein release: an acidic pH and low concentrations of free calcium. The relative contributions of acidic pH and low free calcium to lipoprotein release are not known. Here, we generated fibroblasts that express either normal LDLR or an LDLR variant that is unable to employ the acid-dependent release mechanism to determine the relative contributions of acidic pH and low free calcium on lipoprotein release. We show that endosomal concentrations of free calcium can drive lipoprotein release at rates that are similar to those of acid-dependent release and that the calcium-dependent and acid-dependent mechanisms can cooperate during lipoprotein release. Assessment of lipoprotein uptake by these two cell lines showed that LDL uptake requires the acid-dependent mechanism, while uptake of the VLDL remnant, beta-VLDL, does not. We propose that endosomes use both the acid-dependent and calcium-dependent release mechanisms to drive lipoprotein release and that the acid-dependent process is only required for LDL release.
Collapse
Affiliation(s)
- Zhenze Zhao
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9039, USA
| | | |
Collapse
|
44
|
Russo LC, Goñi CN, Castro LM, Asega AF, Camargo ACM, Trujillo CA, Ulrich H, Glucksman MJ, Scavone C, Ferro ES. Interaction with calmodulin is important for the secretion of thimet oligopeptidase following stimulation. FEBS J 2009; 276:4358-71. [PMID: 19614740 DOI: 10.1111/j.1742-4658.2009.07144.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thimet oligopeptidase (EC 3.4.24.15; EP24.15) was originally described as a neuropeptide-metabolizing enzyme, highly expressed in the brain, kidneys and neuroendocrine tissue. EP24.15 lacks a typical signal peptide sequence for entry into the secretory pathway and is secreted by cells via an unconventional and unknown mechanism. In this study, we identified a novel calcium-dependent interaction between EP24.15 and calmodulin, which is important for the stimulated, but not constitutive, secretion of EP24.15. We demonstrated that, in vitro, EP24.15 and calmodulin physically interact only in the presence of Ca2+, with an estimated Kd value of 0.52 mum. Confocal microscopy confirmed that EP24.15 colocalizes with calmodulin in the cytosol of resting HEK293 cells. This colocalization markedly increases when cells are treated with either the calcium ionophore A23187 or the protein kinase A activator forskolin. Overexpression of calmodulin in HEK293 cells is sufficient to greatly increase the A23187-stimulated secretion of EP24.15, which can be inhibited by the calmodulin inhibitor calmidazolium. The specific inhibition of protein kinase A with KT5720 reduces the A23187-stimulated secretion of EP24.15 and inhibits the synergistic effects of forskolin with A23187. Treatment with calmidazolium and KT5720 nearly abolishes the stimulatory effects of A23187 on EP24.15 secretion. Together, these data suggest that the interaction between EP24.15 and calmodulin is regulated within cells and is important for the stimulated secretion of EP24.15 from HEK293 cells.
Collapse
Affiliation(s)
- Lilian C Russo
- Department of Cell Biology and Development, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Receptor-mediated phagocytosis is a complex process that mediates the internalization, by a cell, of other cells and large particles; this is an important physiological event not only in mammals, but in a wide diversity of organisms. Of simple unicellular organisms that use phagocytosis to extract nutrients, to complex metazoans in which phagocytosis is essential for the innate defence system, as a first line of defence against invading pathogens, as well as for the clearance of damaged, dying or dead cells. Evolution has armed multicellular organisms with a range of receptors expressed on many cells that serve as the molecular basis to bring about phagocytosis, regardless of the organism or the specific physiological event concerned. Key to all phagocytic processes is the finely controlled rearrangement of the actin cytoskeleton, in which Ca(2+) signals play a major role. Ca(2+) is involved in cytoskeletal changes by affecting the actions of a number of contractile proteins, as well as being a cofactor for the activation of a number of intracellular signalling molecules, which are known to play important roles during the initiation, progression and resolution of the phagocytic process. In mammals, the requirement of Ca(2+) for the initial steps in phagocytosis, and the subsequent phagosome maturation, can be quite different depending on the type of cell and on the type of receptor that is driving phagocytosis. In this review we discuss the different receptors that mediate professional and non-professional phagocytosis, and discuss the role of Ca(2+) in the different steps of this complex process.
Collapse
|
46
|
Calábria LK, Garcia Hernandez L, Teixeira RR, Valle de Sousa M, Espindola FS. Identification of calmodulin-binding proteins in brain of worker honeybees. Comp Biochem Physiol B Biochem Mol Biol 2008; 151:41-5. [DOI: 10.1016/j.cbpb.2008.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 05/08/2008] [Accepted: 05/15/2008] [Indexed: 11/28/2022]
|
47
|
The role of calcium and other ions in sorting and delivery in the late endocytic pathway. Biochem Soc Trans 2008; 35:1088-91. [PMID: 17956286 DOI: 10.1042/bst0351088] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The passage of endocytosed receptor-bound ligands and membrane proteins through the endocytic pathway of mammalian cells to lysosomes occurs via early and late endosomes. The latter contain many luminal vesicles and are often referred to as MVBs (multivesicular bodies). The overall morphology of endosomal compartments is, in major part, a consequence of the many fusion events occurring in the endocytic pathway. Kissing events and direct fusion between late endosomes and lysosomes provide a means of delivery to lysosomes. The luminal ionic composition of organelles in the endocytic pathway is of considerable importance both in the trafficking of endocytosed ligands and in the membrane fusion events. In particular, H(+) ions play a role in sorting processes and providing an appropriate environment for the action of lysosomal acid hydrolases. Na(+)/H(+) exchangers in the endosomal membrane have been implicated in the formation of MVBs and sorting into luminal vesicles. Ca(2+) ions are required for fusion events and luminal content condensation in the lysosome. Consistent with an important role for luminal Ca(2+) in traffic through the late endocytic pathway, mutations in the gene encoding mucolipin-1, a lysosomal non-specific cation channel, result in abnormalities in lipid traffic and are associated with the autosomal recessive lysosomal storage disease MLIV (mucolipidosis type IV).
Collapse
|
48
|
Lladó A, Timpson P, Vilà de Muga S, Moretó J, Pol A, Grewal T, Daly RJ, Enrich C, Tebar F. Protein kinase Cdelta and calmodulin regulate epidermal growth factor receptor recycling from early endosomes through Arp2/3 complex and cortactin. Mol Biol Cell 2007; 19:17-29. [PMID: 17959830 DOI: 10.1091/mbc.e07-05-0411] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The intracellular trafficking of the epidermal growth factor receptor (EGFR) is regulated by a cross-talk between calmodulin (CaM) and protein kinase Cdelta (PKCdelta). On inhibition of CaM, PKCdelta promotes the formation of enlarged early endosomes and blocks EGFR recycling and degradation. Here, we show that PKCdelta impairs EGFR trafficking due to the formation of an F-actin coat surrounding early endosomes. The PKCdelta-induced polymerization of actin is orchestrated by the Arp2/3 complex and requires the interaction of cortactin with PKCdelta. Accordingly, inhibition of actin polymerization by using cytochalasin D or by overexpression of active cofilin, restored the normal morphology of the organelle and the recycling of EGFR. Similar results were obtained after down-regulation of cortactin and the sequestration of the Arp2/3 complex. Furthermore we demonstrate an interaction of cortactin with CaM and PKCdelta, the latter being dependent on CaM inhibition. In summary, this study provides the first evidence that CaM and PKCdelta organize actin dynamics in the early endosomal compartment, thereby regulating the intracellular trafficking of EGFR.
Collapse
Affiliation(s)
- Anna Lladó
- Departament de Biologia Cellular, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036-Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Contessi S, Comelli M, Cmet S, Lippe G, Mavelli I. IF(1) distribution in HepG2 cells in relation to ecto-F(0)F (1)ATPsynthase and calmodulin. J Bioenerg Biomembr 2007; 39:291-300. [PMID: 17851741 DOI: 10.1007/s10863-007-9091-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 05/09/2007] [Indexed: 12/01/2022]
Abstract
F(0)F(1)ATPsynthase is now known to be expressed as a plasma membrane receptor for several extracellular ligands. On hepatocytes, ecto-F(0)F(1)ATPsynthase binds apoA-I and triggers HDL endocytosis concomitant with ATP hydrolysis. Considering that inhibitor protein IF(1) was shown to regulate the hydrolytic activity of ecto-F(0)F(1)ATPsynthase and to interact with calmodulin (CaM) in vitro, we investigated the subcellular distributions of IF(1), calmodulin (CaM), OSCP and beta subunits of F(0)F(1)ATPsynthase in HepG2 cells. Using immunofluorescence and Western blotting, we found that around 50% of total cellular IF(1) is localized outside mitochondria, a relevant amount of which is associated to the plasma membrane where we also found Ca(2+)-CaM, OSCP and beta. Confocal microscopy showed that IF(1) colocalized with Ca(2+)-CaM on plasma membrane but not in mitochondria, suggesting that Ca(2+)-CaM may modulate the cell surface availability of IF(1) and thus its ability to inhibit ATP hydrolysis by ecto-F(0)F(1)ATPsynthase. These observations support a hypothesis that the IF(1)-Ca(2+)-CaM complex, forming on plasma membrane, functions in the cellular regulation of HDL endocytosis by hepatocytes.
Collapse
Affiliation(s)
- Stefania Contessi
- Department of Biomedical Sciences and Technologies, MATI Centre of Excellence, University of Udine, Piazzale Kolbe 4, 33100 Udine, Italy
| | | | | | | | | |
Collapse
|
50
|
Saito M, Hanson PI, Schlesinger P. Luminal chloride-dependent activation of endosome calcium channels: patch clamp study of enlarged endosomes. J Biol Chem 2007; 282:27327-27333. [PMID: 17609211 DOI: 10.1074/jbc.m702557200] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Although Ca(2+) release from early endosomes (EE) is important for the fusion of primary endosomes, the presence of an ion channel responsible for releasing calcium from the EE has not been shown. A recent proteomics study has identified the TRPV2 channel protein in EE, suggesting that transient receptor potential-like Ca(2+) channels may be in endosomes. The submicron size of endosomes has made it difficult to study their ion channels in the past. We have overcome this problem by generating enlarged EE with the help of a hydrolysis-deficient SKD1/VPS4B mutant in HEK293 cells. Here we report the first patch clamp recording of a novel endosome calcium channel (ECC) in these enlarged EE. The ECC shows a similar pharmacology to that of the TRPV2 channel. In addition, the ECC has a unique chloride-dependent regulation; it is inhibited by the endosome luminal chloride with a K(50) of 82 mm.
Collapse
Affiliation(s)
- Mitsuyoshi Saito
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110.
| | - Phyllis I Hanson
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - Paul Schlesinger
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| |
Collapse
|