101
|
Lee J, Ye Y. The Roles of Endo-Lysosomes in Unconventional Protein Secretion. Cells 2018; 7:cells7110198. [PMID: 30400277 PMCID: PMC6262434 DOI: 10.3390/cells7110198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Protein secretion in general depends on signal sequence (also named leader sequence), a hydrophobic segment located at or close to the NH2-terminus of a secretory or membrane protein. This sequence guides the entry of nascent polypeptides into the lumen or membranes of the endoplasmic reticulum (ER) for folding, assembly, and export. However, evidence accumulated in recent years has suggested the existence of a collection of unconventional protein secretion (UPS) mechanisms that are independent of the canonical vesicular trafficking route between the ER and the plasma membrane (PM). These UPS mechanisms export soluble proteins bearing no signal sequence. The list of UPS cargos is rapidly expanding, along with the implicated biological functions, but molecular mechanisms accountable for the secretion of leaderless proteins are still poorly defined. This review summarizes our current understanding of UPS mechanisms with an emphasis on the emerging role of endo-lysosomes in this process.
Collapse
Affiliation(s)
- Juhyung Lee
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
102
|
Kühbacher A, Novy K, Quereda JJ, Sachse M, Moya-Nilges M, Wollscheid B, Cossart P, Pizarro-Cerdá J. Listeriolysin O-dependent host surfaceome remodeling modulates Listeria monocytogenes invasion. Pathog Dis 2018; 76:5184460. [PMID: 30445439 PMCID: PMC6282100 DOI: 10.1093/femspd/fty082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Listeria monocytogenes is a pathogenic bacterium that invades epithelial cells by activating host signaling cascades, which promote bacterial engulfment within a phagosome. The pore-forming toxin listeriolysin O (LLO), which is required for bacteria phagosomal escape, has also been associated with the activation of several signaling pathways when secreted by extracellular bacteria, including Ca2+ influx and promotion of L. monocytogenes entry. Quantitative host surfaceome analysis revealed significant quantitative remodeling of a defined set of cell surface glycoproteins upon LLO treatment, including a subset previously identified to play a role in the L. monocytogenes infection process. Our data further shows that the lysosomal-associated membrane proteins LAMP-1 and LAMP-2 are translocated to the cellular surface and those LLO-induced Ca2+ fluxes are required to trigger the surface relocalization of LAMP-1. Finally, we identify late endosomes/lysosomes as the major donor compartments of LAMP-1 upon LLO treatment and by perturbing their function, we suggest that these organelles participate in L. monocytogenes invasion.
Collapse
Affiliation(s)
- Andreas Kühbacher
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France.,INSERM, U604, Paris F-75015, France.,INRA, USC2020, Paris F-75015, France
| | - Karel Novy
- Institute of Molecular Systems Biology and Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Juan J Quereda
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France.,INSERM, U604, Paris F-75015, France.,INRA, USC2020, Paris F-75015, France
| | - Martin Sachse
- Institut Pasteur, UTechS Ultrastructural BioImaging, Paris F-75015, France
| | - Maryse Moya-Nilges
- Institut Pasteur, UTechS Ultrastructural BioImaging, Paris F-75015, France
| | - Bernd Wollscheid
- Institute of Molecular Systems Biology and Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France.,INSERM, U604, Paris F-75015, France.,INRA, USC2020, Paris F-75015, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France.,INSERM, U604, Paris F-75015, France.,INRA, USC2020, Paris F-75015, France.,Institut Pasteur, Unité de Recherche Yersinia, Paris F-75015, France
| |
Collapse
|
103
|
Horn A, Jaiswal JK. Cellular mechanisms and signals that coordinate plasma membrane repair. Cell Mol Life Sci 2018; 75:3751-3770. [PMID: 30051163 PMCID: PMC6541445 DOI: 10.1007/s00018-018-2888-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023]
Abstract
Plasma membrane forms the barrier between the cytoplasm and the environment. Cells constantly and selectively transport molecules across their plasma membrane without disrupting it. Any disruption in the plasma membrane compromises its selective permeability and is lethal, if not rapidly repaired. There is a growing understanding of the organelles, proteins, lipids, and small molecules that help cells signal and efficiently coordinate plasma membrane repair. This review aims to summarize how these subcellular responses are coordinated and how cellular signals generated due to plasma membrane injury interact with each other to spatially and temporally coordinate repair. With the involvement of calcium and redox signaling in single cell and tissue repair, we will discuss how these and other related signals extend from single cell repair to tissue level repair. These signals link repair processes that are activated immediately after plasma membrane injury with longer term processes regulating repair and regeneration of the damaged tissue. We propose that investigating cell and tissue repair as part of a continuum of wound repair mechanisms would be of value in treating degenerative diseases.
Collapse
Affiliation(s)
- Adam Horn
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA.
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
104
|
Grassi S, Chiricozzi E, Mauri L, Sonnino S, Prinetti A. Sphingolipids and neuronal degeneration in lysosomal storage disorders. J Neurochem 2018; 148:600-611. [PMID: 29959861 DOI: 10.1111/jnc.14540] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/27/2022]
Abstract
Ceramide, sphingomyelin, and glycosphingolipids (both neutral and acidic) are characterized by the presence in the lipid moiety of an aliphatic base known as sphingosine. Altogether, they are called sphingolipids and are particularly abundant in neuronal plasma membranes, where, via interactions with the other membrane lipids and membrane proteins, they play a specific role in modulating the cell signaling processes. The metabolic pathways determining the plasma membrane sphingolipid composition are thus the key point for functional changes of the cell properties. Unnatural changes of the neuronal properties are observed in sphingolipidoses, lysosomal storage diseases occurring when a lysosomal sphingolipid hydrolase is not working, leading to the accumulation of the substrate and to its distribution to all the cell membranes interacting with lysosomes. Moreover, secondary accumulation of sphingolipids is a common trait of other lysosomal storage diseases. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
105
|
Abstract
Disruption of plasma membranes is a widespread, common and normal event that occurs in many mechanically challenged tissues (McNeil & Steinhardt, 1997). After injury to the plasma membrane, rapid resealing of the membrane occurs with little loss of intracellular contents.Analysis of plasma membrane repair in the sea urchin egg and early embryos revealed a new model of the mechanism for plasma membrane repair. Resealing of disrupted plasma membranes required external Ca2+ that could be antagonised by Mg2+. Block of Ca2+/calmodulin kinase II, which regulates exocytotic vesicle availability at synapses (Llinás et al., 1991), inhibited membrane resealing. Resealing was also inhibited by botulinum neurotoxins A, B, C1, and tetanus toxin, which disrupt SNARE vesicle docking/fusion proteins. Confocal microscopic observations of exocytotic events in sea urchin eggs and embryos during membrane resealing showed that inhibition of kinesin or myosin motor activity, which are believed to be required for vesicle transport (Goodson et al., 1997), also inhibited membrane resealing and delivery of vesicles to sites of membrane disruption. This pattern of inhibition indicates that membrane repair of micrometre-sized lesions requires vesicle delivery, docking and fusion, similar to the exocytosis of neurotransmitter (Steinhardt et al., 1994; Bi et al., 1995, 1997).The mechanism of resealing in eggs and embyros was found to be a general property of all cells (Steinhardt et al., 1994; Togo et al., 1999). It is now known that elevated intracellular Ca2+ triggers exocytosis in various types of cells (Dan & Poo, 1992; Coorssen et al., 1996), and that endosomal compartments such as lysosomes can behave as Ca2+-regulated exocytotic vesicles (Rodríguez et al., 1997).
Collapse
|
106
|
Kundu ST, Grzeskowiak CL, Fradette JJ, Gibson LA, Rodriguez LB, Creighton CJ, Scott KL, Gibbons DL. TMEM106B drives lung cancer metastasis by inducing TFEB-dependent lysosome synthesis and secretion of cathepsins. Nat Commun 2018; 9:2731. [PMID: 30013069 PMCID: PMC6048095 DOI: 10.1038/s41467-018-05013-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 05/10/2018] [Indexed: 12/19/2022] Open
Abstract
Metastatic lung cancer is the leading cause of cancer-associated mortality worldwide, therefore necessitating novel approaches to identify specific genetic drivers for lung cancer progression and metastasis. We recently performed an in vivo gain-of-function genetic screen to identify driver genes of lung cancer metastasis. In the study reported here, we identify TMEM106B as a primary robust driver of lung cancer metastasis. Ectopic expression of TMEM106B could significantly promote the synthesis of enlarged vesicular lysosomes that are laden with elevated levels of active cathepsins. In a TFEB-dependent manner, TMEM106B could modulate the expression of lysosomal genes of the coordinated lysosomal expression and regulation (CLEAR) pathway in lung cancer cells and patient samples. We also demonstrate that TMEM106B-induced lysosomes undergo calcium-dependent exocytosis, thereby releasing active lysosomal cathepsins necessary for TMEM106B-mediated cancer cell invasion and metastasis in vivo, which could be therapeutically prevented by pharmacological inhibition of cathepsins. Further, in TCGA LUAD data sets, 19% of patients show elevated expression of TMEM106B, which predicts for poor disease-free and overall-survival.
Collapse
Affiliation(s)
- Samrat T Kundu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Caitlin L Grzeskowiak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jared J Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Laura A Gibson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Leticia B Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chad J Creighton
- Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
107
|
Bioorthogonal Conjugation Directed by a Sugar-Sorting Pathway for Continual Tracking of Stressed Organelles. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201802972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
108
|
Xue Z, Zhang E, Liu J, Han J, Han S. Bioorthogonal Conjugation Directed by a Sugar-Sorting Pathway for Continual Tracking of Stressed Organelles. Angew Chem Int Ed Engl 2018; 57:10096-10101. [DOI: 10.1002/anie.201802972] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Zhongwei Xue
- State Key Laboratory for Physical Chemistry of Solid Surfaces; Department of Chemical Biology; College of Chemistry and Chemical Engineering; Key Laboratory for Chemical Biology of Fujian Province; MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, and Innovation Center for Cell Signaling Network; Xiamen University; Xiamen 361005 China
| | - Enkang Zhang
- State Key Laboratory for Physical Chemistry of Solid Surfaces; Department of Chemical Biology; College of Chemistry and Chemical Engineering; Key Laboratory for Chemical Biology of Fujian Province; MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, and Innovation Center for Cell Signaling Network; Xiamen University; Xiamen 361005 China
| | - Jian Liu
- State Key Laboratory for Physical Chemistry of Solid Surfaces; Department of Chemical Biology; College of Chemistry and Chemical Engineering; Key Laboratory for Chemical Biology of Fujian Province; MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, and Innovation Center for Cell Signaling Network; Xiamen University; Xiamen 361005 China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology; Innovation Center for Cell Signaling Network; School of Life Sciences; Xiamen University; Xiamen 361005 China
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid Surfaces; Department of Chemical Biology; College of Chemistry and Chemical Engineering; Key Laboratory for Chemical Biology of Fujian Province; MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, and Innovation Center for Cell Signaling Network; Xiamen University; Xiamen 361005 China
| |
Collapse
|
109
|
Ridinger J, Koeneke E, Kolbinger FR, Koerholz K, Mahboobi S, Hellweg L, Gunkel N, Miller AK, Peterziel H, Schmezer P, Hamacher-Brady A, Witt O, Oehme I. Dual role of HDAC10 in lysosomal exocytosis and DNA repair promotes neuroblastoma chemoresistance. Sci Rep 2018; 8:10039. [PMID: 29968769 PMCID: PMC6030077 DOI: 10.1038/s41598-018-28265-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/15/2018] [Indexed: 12/19/2022] Open
Abstract
Drug resistance is a leading cause for treatment failure in many cancers, including neuroblastoma, the most common solid extracranial childhood malignancy. Previous studies from our lab indicate that histone deacetylase 10 (HDAC10) is important for the homeostasis of lysosomes, i.e. acidic vesicular organelles involved in the degradation of various biomolecules. Here, we show that depleting or inhibiting HDAC10 results in accumulation of lysosomes in chemotherapy-resistant neuroblastoma cell lines, as well as in the intracellular accumulation of the weakly basic chemotherapeutic doxorubicin within lysosomes. Interference with HDAC10 does not block doxorubicin efflux from cells via P-glycoprotein inhibition, but rather via inhibition of lysosomal exocytosis. In particular, intracellular doxorubicin does not remain trapped in lysosomes but also accumulates in the nucleus, where it promotes neuroblastoma cell death. Our data suggest that lysosomal exocytosis under doxorubicin treatment is important for cell survival and that inhibition of HDAC10 further induces DNA double-strand breaks (DSBs), providing additional mechanisms that sensitize neuroblastoma cells to doxorubicin. Taken together, we demonstrate that HDAC10 inhibition in combination with doxorubicin kills neuroblastoma, but not non-malignant cells, both by impeding drug efflux and enhancing DNA damage, providing a novel opportunity to target chemotherapy resistance.
Collapse
Affiliation(s)
- Johannes Ridinger
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Emily Koeneke
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,University of Heidelberg, Heidelberg, Germany
| | - Fiona R Kolbinger
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Katharina Koerholz
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Lars Hellweg
- Research Group Cancer Drug Development, German Cancer Research Center, Heidelberg, Germany
| | - Nikolas Gunkel
- Research Group Cancer Drug Development, German Cancer Research Center, Heidelberg, Germany
| | - Aubry K Miller
- Research Group Cancer Drug Development, German Cancer Research Center, Heidelberg, Germany
| | - Heike Peterziel
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Peter Schmezer
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Anne Hamacher-Brady
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, United States
| | - Olaf Witt
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ina Oehme
- Preclinical Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany. .,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
110
|
Metabolically Activated Adipose Tissue Macrophages Perform Detrimental and Beneficial Functions during Diet-Induced Obesity. Cell Rep 2018; 20:3149-3161. [PMID: 28954231 DOI: 10.1016/j.celrep.2017.08.096] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/25/2017] [Accepted: 08/29/2017] [Indexed: 01/08/2023] Open
Abstract
During obesity, adipose tissue macrophages (ATMs) adopt a metabolically activated (MMe) phenotype. However, the functions of MMe macrophages are poorly understood. Here, we combine proteomic and functional methods to demonstrate that, in addition to potentiating inflammation, MMe macrophages promote dead adipocyte clearance through lysosomal exocytosis. We identify NADPH oxidase 2 (NOX2) as a driver of the inflammatory and adipocyte-clearing properties of MMe macrophages and show that, compared to wild-type, Nox2-/- mice exhibit a time-dependent metabolic phenotype during diet-induced obesity. After 8 weeks of high-fat feeding, Nox2-/- mice exhibit attenuated ATM inflammation and mildly improved glucose tolerance. After 16 weeks of high-fat feeding, Nox2-/- mice develop severe insulin resistance, hepatosteatosis, and visceral lipoatrophy characterized by dead adipocyte accumulation and defective ATM lysosomal exocytosis, a phenotype reproduced in myeloid cell-specific Nox2-/- mice. Collectively, our findings suggest that MMe macrophages perform detrimental and beneficial functions whose contribution to metabolic phenotypes during obesity is determined by disease progression.
Collapse
|
111
|
Ireton K, Van Ngo H, Bhalla M. Interaction of microbial pathogens with host exocytic pathways. Cell Microbiol 2018; 20:e12861. [PMID: 29797532 DOI: 10.1111/cmi.12861] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/13/2018] [Indexed: 02/06/2023]
Abstract
Many microbial pathogens co-opt or perturb host membrane trafficking pathways. This review covers recent examples in which microbes interact with host exocytosis, the fusion of intracellular vesicles with the plasma membrane. The bacterial pathogens Listeria monocytogenes and Staphylococcus aureus subvert recycling endosomal pathways of exocytosis in order to induce their entry into human cells. By contrast, entry of the protozoan pathogen Trypanosoma cruzi or the virus adenovirus into host cells involves exploitation of lysosomal exocytosis. Toxins produced by Bacillus anthracis or Vibrio cholerae interfere with exocytosis pathways mediated by the GTPase Rab11 and the exocyst complex. By doing so, anthrax or cholera toxins impair recycling of cadherins to cell-cell junctions and disrupt the barrier properties of endothelial cells or intestinal epithelial cells, respectively. Uropathogenic Escherichia coli (UPEC) is expelled from bladder epithelial cells through two different exocytic routes that involve sensing of bacteria in vacuoles by host Toll-like receptor 4 (TLR4) or monitoring of the pH of lysosomes harbouring UPEC. The TLR4 pathway is mediated by multiple Rab GTPases and the exocyst, whereas the other pathway involves exocytosis of lysosomes. Expulsion of UPEC through these pathways is thought to benefit the host.
Collapse
Affiliation(s)
- Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Hoan Van Ngo
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
112
|
Dörsam B, Reiners KS, von Strandmann EP. Cancer-derived extracellular vesicles: friend and foe of tumour immunosurveillance. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0481. [PMID: 29158311 PMCID: PMC5717436 DOI: 10.1098/rstb.2016.0481] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2017] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are important players of intercellular signalling mechanisms, including communication with and among immune cells. EVs can affect the surrounding tissue as well as peripheral cells. Recently, EVs have been identified to be involved in the aetiology of several diseases, including cancer. Tumour cell-released EVs or exosomes have been shown to promote a tumour-supporting environment in non-malignant tissue and, thus, benefit metastasis. The underlying mechanisms are numerous: loss of antigen expression, direct suppression of immune effector cells, exchange of nucleic acids, alteration of the recipient cells' transcription and direct suppression of immune cells. Consequently, tumour cells can subvert the host's immune detection as well as suppress the immune system. On the contrary, recent studies reported the existence of EVs able to activate immune cells, thus promoting the tumour-directed immune response. In this article, the immunosuppressive capabilities of EVs, on the one hand, and their potential use in immunoactivation and therapeutic potential, on the other hand, are discussed. This article is part of the discussion meeting issue ‘Extracellular vesicles and the tumour microenvironment’.
Collapse
Affiliation(s)
- Bastian Dörsam
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Hans-Meerwein-Street 3, 35043 Marburg, Germany
| | - Kathrin S Reiners
- Institute of Clinical Chemistry and Clinical Pharmacology, Biomedical Center, University Hospital, University of Bonn, Sigmund-Freud-Street 25, 53127 Bonn, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Hans-Meerwein-Street 3, 35043 Marburg, Germany
| |
Collapse
|
113
|
Etxaniz A, González-Bullón D, Martín C, Ostolaza H. Membrane Repair Mechanisms against Permeabilization by Pore-Forming Toxins. Toxins (Basel) 2018; 10:E234. [PMID: 29890730 PMCID: PMC6024578 DOI: 10.3390/toxins10060234] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 01/29/2023] Open
Abstract
Permeabilization of the plasma membrane represents an important threat for any cell, since it compromises its viability by disrupting cell homeostasis. Numerous pathogenic bacteria produce pore-forming toxins that break plasma membrane integrity and cause cell death by colloid-osmotic lysis. Eukaryotic cells, in turn, have developed different ways to cope with the effects of such membrane piercing. Here, we provide a short overview of the general mechanisms currently proposed for plasma membrane repair, focusing more specifically on the cellular responses to membrane permeabilization by pore-forming toxins and presenting new data on the effects and cellular responses to the permeabilization by an RTX (repeats in toxin) toxin, the adenylate cyclase toxin-hemolysin secreted by the whooping cough bacterium Bordetella pertussis, which we have studied in the laboratory.
Collapse
Affiliation(s)
- Asier Etxaniz
- Biofisika Institute (UPV/EHU, CSIC) and University of the Basque Country (UPV/EHU) Parque Científico s/n, 48940 Leioa, Spain.
| | - David González-Bullón
- Biofisika Institute (UPV/EHU, CSIC) and University of the Basque Country (UPV/EHU) Parque Científico s/n, 48940 Leioa, Spain.
| | - César Martín
- Biofisika Institute (UPV/EHU, CSIC) and University of the Basque Country (UPV/EHU) Parque Científico s/n, 48940 Leioa, Spain.
| | - Helena Ostolaza
- Biofisika Institute (UPV/EHU, CSIC) and University of the Basque Country (UPV/EHU) Parque Científico s/n, 48940 Leioa, Spain.
| |
Collapse
|
114
|
Zhitomirsky B, Assaraf YG. Lysosomal accumulation of anticancer drugs triggers lysosomal exocytosis. Oncotarget 2018; 8:45117-45132. [PMID: 28187461 PMCID: PMC5542171 DOI: 10.18632/oncotarget.15155] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/24/2017] [Indexed: 12/20/2022] Open
Abstract
We have recently shown that hydrophobic weak base anticancer drugs are highly sequestered in acidic lysosomes, inducing TFEB-mediated lysosomal biogenesis and markedly increased lysosome numbers per cell. This enhanced lysosomal sequestration of chemotherapeutics, away from their intracellular targets, provoked cancer multidrug resistance. However, little is known regarding the fate of lysosome-sequestered drugs. While we suggested that sequestered drugs might be expelled from cancer cells via lysosomal exocytosis, no actual drug-induced lysosomal exocytosis was demonstrated. By following the subcellular localization of lysosomes during exposure to lysosomotropic chemotherapeutics, we herein demonstrate that lysosomal drug accumulation results in translocation of lysosomes from the perinuclear zone towards the plasma membrane via movement on microtubule tracks. Furthermore, following translocation to the plasma membrane in drug-treated cells, lysosomes fused with the plasma membrane and released their cargo to the extracellular milieu, as also evidenced by increased levels of the lysosomal enzyme cathepsin D in the extracellular milieu. These findings suggest that lysosomal exocytosis of chemotherapeutic drug-loaded lysosomes is a crucial component of lysosome-mediated cancer multidrug resistance. We further argue that drug-induced lysosomal exocytosis bears important implications on tumor progression, as several lysosomal enzymes were found to play a key role in tumor cell invasion, angiogenesis and metastasis.
Collapse
Affiliation(s)
- Benny Zhitomirsky
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
115
|
Sipos A, Kim KJ, Chow RH, Flodby P, Borok Z, Crandall ED. Alveolar epithelial cell processing of nanoparticles activates autophagy and lysosomal exocytosis. Am J Physiol Lung Cell Mol Physiol 2018; 315:L286-L300. [PMID: 29722567 DOI: 10.1152/ajplung.00108.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Using confocal microscopy, we quantitatively assessed uptake, processing, and egress of near-infrared (NIR)-labeled carboxylated polystyrene nanoparticles (PNP) in live alveolar epithelial cells (AEC) during interactions with primary rat AEC monolayers (RAECM). PNP fluorescence intensity (content) and colocalization with intracellular vesicles in a cell were determined over the entire cell volume via z stacking. Isotropic cuvette-based microfluorimetry was used to determine PNP concentration ([PNP]) from anisotropic measurements of PNP content assessed by confocal microscopy. Results showed that PNP uptake kinetics and steady-state intracellular content decreased as diameter increased from 20 to 200 nm. For 20-nm PNP, uptake rate and steady-state intracellular content increased with increased apical [PNP] but were unaffected by inhibition of endocytic pathways. Intracellular PNP increasingly colocalized with autophagosomes and/or lysosomes over time. PNP egress exhibited fast Ca2+ concentration-dependent release and a slower diffusion-like process. Inhibition of microtubule polymerization curtailed rapid PNP egress, resulting in elevated vesicular and intracellular PNP content. Interference with autophagosome formation led to slower PNP uptake and markedly decreased steady-state intracellular content. At steady state, cytosolic [PNP] was higher than apical [PNP], and vesicular [PNP] (~80% of intracellular PNP content) exceeded both cytosolic and intracellular [PNP]. These data are consistent with the following hypotheses: 1) autophagic processing of nanoparticles is essential for maintenance of AEC integrity; 2) altered autophagy and/or lysosomal exocytosis may lead to AEC injury; and 3) intracellular [PNP] in AEC can be regulated, suggesting strategies for enhancement of nanoparticle-driven AEC gene/drug delivery and/or amelioration of AEC nanoparticle-related cellular toxicity.
Collapse
Affiliation(s)
- Arnold Sipos
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Kwang-Jin Kim
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , Los Angeles, California.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Robert H Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Per Flodby
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zea Borok
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Edward D Crandall
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pathology, Keck School of Medicine, University of Southern California , Los Angeles, California.,Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| |
Collapse
|
116
|
Liu CL, Guo J, Zhang X, Sukhova GK, Libby P, Shi GP. Cysteine protease cathepsins in cardiovascular disease: from basic research to clinical trials. Nat Rev Cardiol 2018; 15:351-370. [DOI: 10.1038/s41569-018-0002-3] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
117
|
Michelet X, Tuli A, Gan H, Geadas C, Sharma M, Remold HG, Brenner MB. Lysosome-Mediated Plasma Membrane Repair Is Dependent on the Small GTPase Arl8b and Determines Cell Death Type in Mycobacterium tuberculosis Infection. THE JOURNAL OF IMMUNOLOGY 2018; 200:3160-3169. [PMID: 29592961 DOI: 10.4049/jimmunol.1700829] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 02/23/2018] [Indexed: 01/06/2023]
Abstract
Mycobacterium tuberculosis is an extremely successful pathogen, and its success is widely attributed to its ability to manipulate the intracellular environment of macrophages. A central phenomenon of tuberculosis pathology enabling immune evasion is the capacity of virulent M. tuberculosis (H37Rv) to induce macrophage necrosis, which facilitates the escape of the mycobacteria from the macrophage and spread of infection. In contrast, avirulent M. tuberculosis (H37Ra) induces macrophage apoptosis, which permits Ag presentation and activation of adaptive immunity. Previously, we found that H37Rv induces plasma membrane microdisruptions, leading to necrosis in the absence of plasma membrane repair. In contrast, H37Ra permits plasma membrane repair, which changes the host cell death modality to apoptosis, suggesting that membrane repair is critical for sequestering the pathogen in apoptotic vesicles. However, mechanisms of plasma membrane repair induced in response to M. tuberculosis infection remain unknown. Plasma membrane repair is known to induce a Ca2+-mediated signaling, which recruits lysosomes to the area of damaged plasma membrane sites for its resealing. In this study, we found that the small GTPase Arl8b is required for plasma membrane repair by controlling the exocytosis of lysosomes in cell lines and in human primary macrophages. Importantly, we found that the Arl8b secretion pathway is crucial to control the type of cell death of the M. tuberculosis-infected macrophages. Indeed, Arl8b-depleted macrophages infected with avirulent H37Ra undergo necrotic instead of apoptotic cell death. These findings suggest that membrane repair mediated by Arl8b may be an important mechanism distinguishing avirulent from virulent M. tuberculosis-induced necrotic cell death.
Collapse
Affiliation(s)
- Xavier Michelet
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115;
| | - Amit Tuli
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; .,Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Huixian Gan
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Carolina Geadas
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Mahak Sharma
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab 140306, India
| | - Heinz G Remold
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Michael B Brenner
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
118
|
Sterea AM, Almasi S, El Hiani Y. The hidden potential of lysosomal ion channels: A new era of oncogenes. Cell Calcium 2018; 72:91-103. [PMID: 29748137 DOI: 10.1016/j.ceca.2018.02.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/28/2018] [Accepted: 02/28/2018] [Indexed: 01/14/2023]
Abstract
Lysosomes serve as the control centre for cellular clearance. These membrane-bound organelles receive biomolecules destined for degradation from intracellular and extracellular pathways; thus, facilitating the production of energy and shaping the fate of the cell. At the base of their functionality are the lysosomal ion channels which mediate the function of the lysosome through the modulation of ion influx and efflux. Ion channels form pores in the membrane of lysosomes and allow the passage of ions, a seemingly simple task which harbours the potential of overthrowing the cell's stability. Considered the master regulators of ion homeostasis, these integral membrane proteins enable the proper operation of the lysosome. Defects in the structure or function of these ion channels lead to the development of lysosomal storage diseases, neurodegenerative diseases and cancer. Although more than 50 years have passed since their discovery, lysosomes are not yet fully understood, with their ion channels being even less well characterized. However, significant improvements have been made in the development of drugs targeted against these ion channels as a means of combating diseases. In this review, we will examine how Ca2+, K+, Na+ and Cl- ion channels affect the function of the lysosome, their involvement in hereditary and spontaneous diseases, and current ion channel-based therapies.
Collapse
Affiliation(s)
- Andra M Sterea
- Departments of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shekoufeh Almasi
- Departments of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yassine El Hiani
- Departments of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
119
|
Verweij FJ, Bebelman MP, Jimenez CR, Garcia-Vallejo JJ, Janssen H, Neefjes J, Knol JC, de Goeij-de Haas R, Piersma SR, Baglio SR, Verhage M, Middeldorp JM, Zomer A, van Rheenen J, Coppolino MG, Hurbain I, Raposo G, Smit MJ, Toonen RFG, van Niel G, Pegtel DM. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J Cell Biol 2018; 217:1129-1142. [PMID: 29339438 PMCID: PMC5839777 DOI: 10.1083/jcb.201703206] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 10/18/2017] [Accepted: 12/01/2017] [Indexed: 11/22/2022] Open
Abstract
All mammalian cells release small endosome-derived exosomes that function in intercellular communication, but the secretion process is poorly understood. Verweij et al. developed a live-imaging approach and demonstrate that external cues can trigger exosome release from a subpopulation of multivesicular bodies by phosphorylating the target membrane SNARE SNAP23 at serine residue 110. Exosomes are small endosome-derived extracellular vesicles implicated in cell–cell communication and are secreted by living cells when multivesicular bodies (MVBs) fuse with the plasma membrane (PM). Current techniques to study exosome physiology are based on isolation procedures after secretion, precluding direct and dynamic insight into the mechanics of exosome biogenesis and the regulation of their release. In this study, we propose real-time visualization of MVB–PM fusion to overcome these limitations. We designed tetraspanin-based pH-sensitive optical reporters that detect MVB–PM fusion using live total internal reflection fluorescence and dynamic correlative light–electron microscopy. Quantitative analysis demonstrates that MVB–PM fusion frequency is reduced by depleting the target membrane SNAREs SNAP23 and syntaxin-4 but also can be induced in single cells by stimulation of the histamine H1 receptor (H1HR). Interestingly, activation of H1R1 in HeLa cells increases Ser110 phosphorylation of SNAP23, promoting MVB–PM fusion and the release of CD63-enriched exosomes. Using this single-cell resolution approach, we highlight the modulatory dynamics of MVB exocytosis that will help to increase our understanding of exosome physiology and identify druggable targets in exosome-associated pathologies.
Collapse
Affiliation(s)
- Frederik Johannes Verweij
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands .,Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris, France
| | - Maarten P Bebelman
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands.,Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, Netherlands
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Hans Janssen
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jacques Neefjes
- Department of Chemical Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Jaco C Knol
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Richard de Goeij-de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Sander R Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - S Rubina Baglio
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Matthijs Verhage
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Jaap M Middeldorp
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Anoek Zomer
- Cancer Genomics Netherlands-Hubrecht Institute-Koninklijke Nederlandse Akademie van Wetenschappen, Utrecht, Netherlands.,University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jacco van Rheenen
- Cancer Genomics Netherlands-Hubrecht Institute-Koninklijke Nederlandse Akademie van Wetenschappen, Utrecht, Netherlands.,University Medical Centre Utrecht, Utrecht, Netherlands
| | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Ilse Hurbain
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris, France
| | - Graça Raposo
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris, France
| | - Martine J Smit
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, Netherlands
| | - Ruud F G Toonen
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Guillaume van Niel
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Centre National de la Recherché Scientifique, UMR 144, Paris, France.,Cell and Tissue Imaging Core Facility PICT-IBiSA, Institut Curie, Paris, France
| | - D Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
120
|
Lee JJA, Maruyama R, Sakurai H, Yokota T. Cell Membrane Repair Assay Using a Two-photon Laser Microscope. J Vis Exp 2018. [PMID: 29364240 DOI: 10.3791/56999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Numerous pathophysiological insults can cause damage to cell membranes and, when coupled with innate defects in cell membrane repair or integrity, can result in disease. Understanding the underlying molecular mechanisms surrounding cell membrane repair is, therefore, an important objective to the development of novel therapeutic strategies for diseases associated with dysfunctional cell membrane dynamics. Many in vitro and in vivo studies aimed at understanding cell membrane resealing in various disease contexts utilize two-photon laser ablation as a standard for determining functional outcomes following experimental treatments. In this assay, cell membranes are subjected to wounding with a two-photon laser, which causes the cell membrane to rupture and fluorescent dye to infiltrate the cell. The intensity of fluorescence within the cell can then be monitored to quantify the cell's ability to reseal itself. There are several alternative methods for assessing cell membrane response to injury, as well as great variation in the two-photon laser wounding approach itself, therefore, a single, unified model of cell wounding would beneficially serve to decrease the variation between these methodologies. In this article, we outline a simple two-photon laser wounding protocol for assessing cell membrane repair in vitro in both healthy and dysferlinopathy patient fibroblast cells transfected with or without a full-length dysferlin plasmid.
Collapse
Affiliation(s)
- Joshua J A Lee
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | | | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry;
| |
Collapse
|
121
|
Naegeli KM, Hastie E, Garde A, Wang Z, Keeley DP, Gordon KL, Pani AM, Kelley LC, Morrissey MA, Chi Q, Goldstein B, Sherwood DR. Cell Invasion In Vivo via Rapid Exocytosis of a Transient Lysosome-Derived Membrane Domain. Dev Cell 2017; 43:403-417.e10. [PMID: 29161591 DOI: 10.1016/j.devcel.2017.10.024] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/19/2017] [Accepted: 10/22/2017] [Indexed: 11/27/2022]
Abstract
Invasive cells use small invadopodia to breach basement membrane (BM), a dense matrix that encases tissues. Following the breach, a large protrusion forms to clear a path for tissue entry by poorly understood mechanisms. Using RNAi screening for defects in Caenorhabditis elegans anchor cell (AC) invasion, we found that UNC-6(netrin)/UNC-40(DCC) signaling at the BM breach site directs exocytosis of lysosomes using the exocyst and SNARE SNAP-29 to form a large protrusion that invades vulval tissue. Live-cell imaging revealed that the protrusion is enriched in the matrix metalloprotease ZMP-1 and transiently expands AC volume by more than 20%, displacing surrounding BM and vulval epithelium. Photobleaching and genetic perturbations showed that the BM receptor dystroglycan forms a membrane diffusion barrier at the neck of the protrusion, which enables protrusion growth. Together these studies define a netrin-dependent pathway that builds an invasive protrusion, an isolated lysosome-derived membrane structure specialized to breach tissue barriers.
Collapse
Affiliation(s)
- Kaleb M Naegeli
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708, USA
| | - Eric Hastie
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Aastha Garde
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Kacy L Gordon
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Ariel M Pani
- Biology Department and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Laura C Kelley
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Meghan A Morrissey
- The Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Qiuyi Chi
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - Bob Goldstein
- Biology Department and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
122
|
Villeneuve J, Bassaganyas L, Lepreux S, Chiritoiu M, Costet P, Ripoche J, Malhotra V, Schekman R. Unconventional secretion of FABP4 by endosomes and secretory lysosomes. J Cell Biol 2017; 217:649-665. [PMID: 29212659 PMCID: PMC5800802 DOI: 10.1083/jcb.201705047] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 01/30/2023] Open
Abstract
Adipocytes secrete fatty acid binding protein 4, which influences glucose production in hepatocytes and insulin secretion in pancreatic β-cells, but the mechanisms of its secretion are unclear. Villeneuve et al. show that FABP4 is secreted unconventionally through enclosure within endosomes and secretory lysosomes. An appreciation of the functional properties of the cytoplasmic fatty acid binding protein 4 (FABP4) has advanced with the recent demonstration that an extracellular form secreted by adipocytes regulates a wide range of physiological functions. Little, however, is known about the mechanisms that mediate the unconventional secretion of FABP4. Here, we demonstrate that FABP4 secretion is mediated by a membrane-bounded compartment, independent of the conventional endoplasmic reticulum–Golgi secretory pathway. We show that FABP4 secretion is also independent of GRASP proteins, autophagy, and multivesicular bodies but involves enclosure within endosomes and secretory lysosomes. We highlight the physiological significance of this pathway with the demonstration that an increase in plasma levels of FABP4 is inhibited by chloroquine treatment of mice. These findings chart the pathway of FABP4 secretion and provide a potential therapeutic means to control metabolic disorders associated with its dysregulated secretion.
Collapse
Affiliation(s)
- Julien Villeneuve
- Center for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Molecular and Cell Biology and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA
| | - Laia Bassaganyas
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
| | - Sebastien Lepreux
- Institut National de la Santé et de la Recherche Médicale U1026, Université de Bordeaux, Bordeaux, France
| | - Marioara Chiritoiu
- Center for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Pierre Costet
- Service des Animaleries, Université de Bordeaux, Bordeaux, France
| | - Jean Ripoche
- Institut National de la Santé et de la Recherche Médicale U1026, Université de Bordeaux, Bordeaux, France
| | - Vivek Malhotra
- Center for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain .,Universitat Pompeu Fabra, Barcelona, Spain.,Institutio Catalana de Recerca i Estudis Avancats, Barcelona, Spain
| | - Randy Schekman
- Department of Molecular and Cell Biology and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
123
|
Padamsey Z, McGuinness L, Emptage NJ. Inhibition of lysosomal Ca 2+ signalling disrupts dendritic spine structure and impairs wound healing in neurons. Commun Integr Biol 2017; 10:e1344802. [PMID: 29259727 PMCID: PMC5731510 DOI: 10.1080/19420889.2017.1344802] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 11/21/2022] Open
Abstract
A growing body of evidence suggests that lysosomes, which have traditionally been regarded as degradative organelles, can function as Ca2+ stores, regulated by the second messenger nicotinic acid adenine dinucleotide phosphate (NAADP). We previously demonstrated that in hippocampal pyramidal neurons, activity-dependent Ca2+ release from these stores triggers fusion of the lysosome with the plasma membrane. We found that the physiological role of this Ca2+-dependent fusion was to maintain the long-term structural enlargement of dendritic spines induced by synaptic activity. Here, we examined the pathophysiological consequences of lysosomal dysfunction in hippocampal pyramidal neurons by chronically inhibiting lysosomal Ca2+ signalling using the NAADP antagonist, NED-19. We found that within just 20 hours, inhibition of lysosomal function led to a profound intracellular accumulation of lysosomal membrane. This was accompanied by a significant change in dendritic spine structure, which included a lengthening of dendritic spines, an increase in the number of filipodia, and an overall decrease in spine number. Inhibition of lysosomal function also inhibited wound healing in neurons by preventing lysosomal fusion with the plasma membrane. Neurons were therefore more susceptible to injury. Our findings suggest that dysfunction in lysosomal Ca2+ signalling and lysosomal fusion with the plasma membrane may contribute to the loss of dendritic spines and neurons seen in neurological disorders, such as Niemann-Pick disease type C1, in which lysosomal function is impaired.
Collapse
Affiliation(s)
- Zahid Padamsey
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Nigel J Emptage
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
124
|
Pereira GJS, Antonioli M, Hirata H, Ureshino RP, Nascimento AR, Bincoletto C, Vescovo T, Piacentini M, Fimia GM, Smaili SS. Glutamate induces autophagy via the two-pore channels in neural cells. Oncotarget 2017; 8:12730-12740. [PMID: 28055974 PMCID: PMC5355049 DOI: 10.18632/oncotarget.14404] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/27/2016] [Indexed: 12/22/2022] Open
Abstract
NAADP (nicotinic acid adenine dinucleotide phosphate) has been proposed as a second messenger for glutamate in neuronal and glial cells via the activation of the lysosomal Ca2+ channels TPC1 and TPC2. However, the activities of glutamate that are mediated by NAADP remain unclear. In this study, we evaluated the effect of glutamate on autophagy in astrocytes at physiological, non-toxic concentration. We found that glutamate induces autophagy at similar extent as NAADP. By contrast, the NAADP antagonist NED-19 or SiRNA-mediated inhibition of TPC1/2 decreases autophagy induced by glutamate, confirming a role for NAADP in this pathway. The involvement of TPC1/2 in glutamate-induced autophagy was also confirmed in SHSY5Y neuroblastoma cells. Finally, we show that glutamate leads to a NAADP-dependent activation of AMPK, which is required for autophagy induction, while mTOR activity is not affected by this treatment. Taken together, our results indicate that glutamate stimulates autophagy via NAADP/TPC/AMPK axis, providing new insights of how Ca2+ signalling glutamate-mediated can control the cell metabolism in the central nervous system.
Collapse
Affiliation(s)
- Gustavo J S Pereira
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Manuela Antonioli
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.,Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy
| | - Hanako Hirata
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Rodrigo P Ureshino
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Aline R Nascimento
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Claudia Bincoletto
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Tiziana Vescovo
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.,Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy
| | - Gian Maria Fimia
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy.,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Soraya S Smaili
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| |
Collapse
|
125
|
Chakraborty K, Leung K, Krishnan Y. High lumenal chloride in the lysosome is critical for lysosome function. eLife 2017; 6. [PMID: 28742019 PMCID: PMC5526669 DOI: 10.7554/elife.28862] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 06/23/2017] [Indexed: 12/31/2022] Open
Abstract
Lysosomes are organelles responsible for the breakdown and recycling of cellular machinery. Dysfunctional lysosomes give rise to lysosomal storage disorders as well as common neurodegenerative diseases. Here, we use a DNA-based, fluorescent chloride reporter to measure lysosomal chloride in Caenorhabditis elegans as well as murine and human cell culture models of lysosomal diseases. We find that the lysosome is highly enriched in chloride, and that chloride reduction correlates directly with a loss in the degradative function of the lysosome. In nematodes and mammalian cell culture models of diverse lysosomal disorders, where previously only lysosomal pH dysregulation has been described, massive reduction of lumenal chloride is observed that is ~103 fold greater than the accompanying pH change. Reducing chloride within the lysosome impacts Ca2+ release from the lysosome and impedes the activity of specific lysosomal enzymes indicating a broader role for chloride in lysosomal function. DOI:http://dx.doi.org/10.7554/eLife.28862.001 In cells, worn out proteins and other unnecessary materials are sent to small compartments called lysosomes to be broken down and recycled. Lysosomes contain many different proteins including some that break down waste material into recyclable fragments and others that transport the fragments out of the lysosome. If any of these proteins do not work, waste products build up and cause disease. There are around 70 such lysosomal storage diseases, each arising from a different lysosomal protein not working correctly. A recently developed “nanodevice” called Clensor can measure the levels of chloride ions inside cells. Clensor is constructed from DNA, and its fluorescence changes when it detects chloride ions. Although chloride ions have many biological roles, chloride ion levels had not been measured inside a living organism. Now, Chakraborty et al. – including some of the researchers who developed Clensor – have used this nanodevice to examine chloride ion levels in the lysosomes of the roundworm Caenorhabditis elegans. This revealed that the lysosomes contain high levels of chloride ions. Furthermore, reducing the amount of chloride in the lysosomes made them worse at breaking down waste. Do lysosomes affected by lysosome storage diseases also contain low levels of chloride ions? To find out, Chakraborty et al. used Clensor to study C. elegans worms and mouse and human cells whose lysosomes accumulate waste products. In all these cases, the levels of chloride in the diseased lysosomes were much lower than normal. This had a number of effects on how the lysosomes worked, such as reducing the activity of key lysosomal proteins. Chakraborty et al. also found that Clensor can be used to distinguish between different lysosomal storage diseases. This means that in the future, Clensor (or similar methods that directly measure chloride ion levels in lysosomes) may be useful not just for research purposes. They may also be valuable for diagnosing lysosomal storage diseases early in infancy that, if left undiagnosed, are fatal. DOI:http://dx.doi.org/10.7554/eLife.28862.002
Collapse
Affiliation(s)
- Kasturi Chakraborty
- Department of Chemistry, University of Chicago, Chicago, United States.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, United States
| | - KaHo Leung
- Department of Chemistry, University of Chicago, Chicago, United States.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, United States
| | - Yamuna Krishnan
- Department of Chemistry, University of Chicago, Chicago, United States.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, United States
| |
Collapse
|
126
|
Nüße J, Blumrich EM, Mirastschijski U, Kappelmann L, Kelm S, Dietz F. Intra- or extra-exosomal secretion of HDGF isoforms: the extraordinary function of the HDGF-A N-terminal peptide. Biol Chem 2017; 398:793-811. [DOI: 10.1515/hsz-2016-0315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022]
Abstract
Abstract
Hepatoma-derived growth factor (HDGF) is a protein with diverse intracellular functions. Moreover, after non-conventional secretion, extracellular HDGF is able to influence different signaling pathways, leading for example to induction of processes like epithelial-mesenchymal transition (EMT) and cell migration. Intriguingly, in recent proteome studies, HDGF was also found secreted by special microvesicles called exosomes. Recently, we demonstrated the existence of two new HDGF isoforms (B and C). These isoforms are involved in different cellular processes than HDGF-A. Along this line, in the present study we discovered that full length HDGF-A clearly is located inside of exosomes, whereas the isoforms HDGF-B and HDGF-C are found exclusively on the outer surface. Furthermore, while HDGF-B and HDGF-C seem to use exosomes mediated pathway exclusively, HDGF-A was found also as unbound protein in the conditioned media. The new finding of an intra- or extra-exosomal localisation of protein splice variants opens a fascinating new perspective concerning functional diversity of HDGF isoforms. Dysregulation of HDGF expression during cancer development and tumor progression is a commonly known fact. With our new findings, unraveling the potential functional impact according to physiological versus pathophysiologically altered levels and compositions of intra- and extra-exosomal HDGF has to be addressed in future studies.
Collapse
|
127
|
Di Paola S, Scotto-Rosato A, Medina DL. TRPML1: The Ca (2+)retaker of the lysosome. Cell Calcium 2017; 69:112-121. [PMID: 28689729 DOI: 10.1016/j.ceca.2017.06.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/16/2017] [Accepted: 06/16/2017] [Indexed: 12/27/2022]
Abstract
Efficient functioning of lysosome is necessary to ensure the correct performance of a variety of intracellular processes such as degradation of cargoes coming from the endocytic and autophagic pathways, recycling of organelles, and signaling mechanisms involved in cellular adaptation to nutrient availability. Mutations in lysosomal genes lead to more than 50 lysosomal storage disorders (LSDs). Among them, mutations in the gene encoding TRPML1 (MCOLN1) cause Mucolipidosis type IV (MLIV), a recessive LSD characterized by neurodegeneration, psychomotor retardation, ophthalmologic defects and achlorhydria. At the cellular level, MLIV patient fibroblasts show enlargement and engulfment of the late endo-lysosomal compartment, autophagy impairment, and accumulation of lipids and glycosaminoglycans. TRPML1 is the most extensively studied member of a small family of genes that also includes TRPML2 and TRPML3, and it has been found to participate in vesicular trafficking, lipid and ion homeostasis, and autophagy. In this review we will provide an update on the latest and more novel findings related to the functions of TRPMLs, with particular focus on the emerging role of TRPML1 and lysosomal calcium signaling in autophagy. Moreover, we will also discuss new potential therapeutic approaches for MLIV and LSDs based on the modulation of TRPML1-mediated signaling.
Collapse
Affiliation(s)
- Simone Di Paola
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli ,NA, Italy
| | - Anna Scotto-Rosato
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli ,NA, Italy
| | - Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli ,NA, Italy.
| |
Collapse
|
128
|
Couto NF, Pedersane D, Rezende L, Dias PP, Corbani TL, Bentini LC, Oliveira ACS, Kelles LF, Castro-Gomes T, Andrade LO. LAMP-2 absence interferes with plasma membrane repair and decreases T. cruzi host cell invasion. PLoS Negl Trop Dis 2017; 11:e0005657. [PMID: 28586379 PMCID: PMC5473579 DOI: 10.1371/journal.pntd.0005657] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 06/16/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022] Open
Abstract
Trypanosoma cruzi enters host cells by subverting the mechanism of cell membrane repair. In this process, the parasite induces small injuries in the host cell membrane leading to calcium entry and lysosomal exocytosis, which are followed by compensatory endocytosis events that drive parasites into host cells. We have previously shown that absence of both LAMP-1 and 2, major components of lysosomal membranes, decreases invasion of T. cruzi into host cells, but the mechanism by which they interfere with parasite invasion has not been described. Here we investigated the role of these proteins in parasitophorous vacuole morphology, host cell lysosomal exocytosis, and membrane repair ability. First, we showed that cells lacking only LAMP-2 present the same invasion phenotype as LAMP1/2-/- cells, indicating that LAMP-2 is an important player during T. cruzi invasion process. Second, neither vacuole morphology nor lysosomal exocytosis was altered in LAMP-2 lacking cells (LAMP2-/- and LAMP1/2-/- cells). We then investigated the ability of LAMP-2 deficient cells to perform compensatory endocytosis upon lysosomal secretion, the mechanism by which cells repair their membrane and T. cruzi ultimately enters cells. We observed that these cells perform less endocytosis upon injury when compared to WT cells. This was a consequence of impaired cholesterol traffic in cells lacking LAMP-2 and its influence in the distribution of caveolin-1 at the cell plasma membrane, which is crucial for plasma membrane repair. The results presented here show the major role of LAMP-2 in caveolin traffic and membrane repair and consequently in T. cruzi invasion.
Collapse
Affiliation(s)
| | - Dina Pedersane
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luisa Rezende
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patrícia P. Dias
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tayanne L. Corbani
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lívia C. Bentini
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anny C. S. Oliveira
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ludmila F. Kelles
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thiago Castro-Gomes
- Department of Biochemistry and Immunology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana O. Andrade
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
129
|
Encarnação M, Espada L, Escrevente C, Mateus D, Ramalho J, Michelet X, Santarino I, Hsu VW, Brenner MB, Barral DC, Vieira OV. A Rab3a-dependent complex essential for lysosome positioning and plasma membrane repair. J Cell Biol 2017; 213:631-40. [PMID: 27325790 PMCID: PMC4915190 DOI: 10.1083/jcb.201511093] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/27/2016] [Indexed: 11/22/2022] Open
Abstract
Encarnação et al. show that Rab3a, together with its newly identified effector NMHC IIA, mediates the positioning of peripheral lysosomes in nonsecretory cells, thereby promoting lysosome exocytosis and plasma membrane repair. Lysosome exocytosis plays a major role in resealing plasma membrane (PM) disruptions. This process involves two sequential steps. First, lysosomes are recruited to the periphery of the cell and then fuse with the damaged PM. However, the trafficking molecular machinery involved in lysosome exocytosis and PM repair (PMR) is poorly understood. We performed a systematic screen of the human Rab family to identify Rabs required for lysosome exocytosis and PMR. Rab3a, which partially localizes to peripheral lysosomes, was one of the most robust hits. Silencing of Rab3a or its effector, synaptotagmin-like protein 4a (Slp4-a), leads to the collapse of lysosomes to the perinuclear region and inhibition of PMR. Importantly, we have also identified a new Rab3 effector, nonmuscle myosin heavy chain IIA, as part of the complex formed by Rab3a and Slp4-a that is responsible for lysosome positioning at the cell periphery and lysosome exocytosis.
Collapse
Affiliation(s)
- Marisa Encarnação
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Lília Espada
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Cristina Escrevente
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Denisa Mateus
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - José Ramalho
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Xavier Michelet
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Inês Santarino
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Victor W Hsu
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Duarte C Barral
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Otília V Vieira
- Centro de Estudos de Doenças Crónicas, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| |
Collapse
|
130
|
Afzali AM, Ruck T, Wiendl H, Meuth SG. Animal models in idiopathic inflammatory myopathies: How to overcome a translational roadblock? Autoimmun Rev 2017; 16:478-494. [DOI: 10.1016/j.autrev.2017.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 12/19/2022]
|
131
|
BORC/kinesin-1 ensemble drives polarized transport of lysosomes into the axon. Proc Natl Acad Sci U S A 2017; 114:E2955-E2964. [PMID: 28320970 DOI: 10.1073/pnas.1616363114] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The ability of lysosomes to move within the cytoplasm is important for many cellular functions. This ability is particularly critical in neurons, which comprise vast, highly differentiated domains such as the axon and dendrites. The mechanisms that control lysosome movement in these domains, however, remain poorly understood. Here we show that an ensemble of BORC, Arl8, SKIP, and kinesin-1, previously shown to mediate centrifugal transport of lysosomes in nonneuronal cells, specifically drives lysosome transport into the axon, and not the dendrites, in cultured rat hippocampal neurons. This transport is essential for maintenance of axonal growth-cone dynamics and autophagosome turnover. Our findings illustrate how a general mechanism for lysosome dispersal in nonneuronal cells is adapted to drive polarized transport in neurons, and emphasize the importance of this mechanism for critical axonal processes.
Collapse
|
132
|
Abstract
Lysosomes, the major membrane-bound degradative organelles, have a multitude of functions in eukaryotic cells. Lysosomes are the terminal compartments in the endocytic pathway, though they display highly dynamic behaviors, fusing with each other and with late endosomes in the endocytic pathway, and with the plasma membrane during regulated exocytosis and for wound repair. After fusing with late endosomes, lysosomes are reformed from the resulting hybrid organelles through a process that involves budding of a nascent lysosome, extension of the nascent lysosome from the hybrid organelle, while remaining connected by a membrane bridge, and scission of the membrane bridge to release the newly formed lysosome. The newly formed lysosomes undergo cycles of homotypic fusion and fission reactions to form mature lysosomes. In this study, we used a forward genetic screen in Caenorhabditis elegans to identify six regulators of lysosome biology. We show that these proteins function in different steps of lysosome biology, regulating lysosome formation, lysosome fusion, and lysosome degradation.
Collapse
|
133
|
Abstract
Membrane repair emerges as an innate defense protecting target cells against bacterial pore-forming toxins. Here, we report the first paradigm of Ca2+-dependent repair following attack by a small β-pore-forming toxin, namely, plasmid-encoded phobalysin of Photobacterium damselae subsp. damselae. In striking contrast, Vibrio cholerae cytolysin, the closest ortholog of phobalysin, subverted repair. Mutational analysis uncovered a role of channel width in toxicity and repair. Thus, the replacement of serine at phobalysin´s presumed channel narrow point with the bulkier tryptophan, the corresponding residue in Vibrio cholerae cytolysin (W318), modulated Ca2+ influx, lysosomal exocytosis, and membrane repair. And yet, replacing tryptophan (W318) with serine in Vibrio cholerae cytolysin enhanced toxicity. The data reveal divergent strategies evolved by two related small β-pore-forming toxins to manipulate target cells: phobalysin leads to fulminant perturbation of ion concentrations, closely followed by Ca2+ influx-dependent membrane repair. In contrast, V. cholerae cytolysin causes insidious perturbations and escapes control by the cellular wounded membrane repair-like response. Previous studies demonstrated that large transmembrane pores, such as those formed by perforin or bacterial toxins of the cholesterol-dependent cytolysin family, trigger rapid, Ca2+ influx-dependent repair mechanisms. In contrast, recovery from attack by the small β-pore-forming Staphylococcus aureus alpha-toxin or aerolysin is slow in comparison and does not depend on extracellular Ca2+. To further elucidate the scope of Ca2+ influx-dependent repair and understand its limitations, we compared the cellular responses to phobalysin and V. cholerae cytolysin, two related small β-pore-forming toxins which create membrane pores of slightly different sizes. The data indicate that the channel width of a small β-pore-forming toxin is a critical determinant of both primary toxicity and susceptibility to Ca2+-dependent repair.
Collapse
|
134
|
Rega LR, Polishchuk E, Montefusco S, Napolitano G, Tozzi G, Zhang J, Bellomo F, Taranta A, Pastore A, Polishchuk R, Piemonte F, Medina DL, Catz SD, Ballabio A, Emma F. Activation of the transcription factor EB rescues lysosomal abnormalities in cystinotic kidney cells. Kidney Int 2017; 89:862-73. [PMID: 26994576 DOI: 10.1016/j.kint.2015.12.045] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 12/22/2015] [Accepted: 12/30/2015] [Indexed: 11/15/2022]
Abstract
Nephropathic cystinosis is a rare autosomal recessive lysosomal storage disease characterized by accumulation of cystine into lysosomes secondary to mutations in the cystine lysosomal transporter, cystinosin. The defect initially causes proximal tubular dysfunction (Fanconi syndrome) which in time progresses to end-stage renal disease. Cystinotic patients treated with the cystine-depleting agent, cysteamine, have improved life expectancy, delayed progression to chronic renal failure, but persistence of Fanconi syndrome. Here, we have investigated the role of the transcription factor EB (TFEB), a master regulator of the autophagy-lysosomal pathway, in conditionally immortalized proximal tubular epithelial cells derived from the urine of a healthy volunteer or a cystinotic patient. Lack of cystinosin reduced TFEB expression and induced TFEB nuclear translocation. Stimulation of endogenous TFEB activity by genistein, or overexpression of exogenous TFEB lowered cystine levels within 24 hours in cystinotic cells. Overexpression of TFEB also stimulated delayed endocytic cargo processing within 24 hours. Rescue of other abnormalities of the lysosomal compartment was observed but required prolonged expression of TFEB. These abnormalities could not be corrected with cysteamine. Thus, these data show that the consequences of cystinosin deficiency are not restricted to cystine accumulation and support the role of TFEB as a therapeutic target for the treatment of lysosomal storage diseases, in particular of cystinosis.
Collapse
Affiliation(s)
- Laura R Rega
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy.
| | - Elena Polishchuk
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Sandro Montefusco
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | | | - Giulia Tozzi
- Unit for Muscular and Neurodegenerative Diseases, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Jinzhong Zhang
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Francesco Bellomo
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Anna Taranta
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Anna Pastore
- Laboratory of Proteomics and Metabolomics, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Fiorella Piemonte
- Unit for Muscular and Neurodegenerative Diseases, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Sergio D Catz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Francesco Emma
- Division of Nephrology and Dialysis, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy
| |
Collapse
|
135
|
Borland H, Vilhardt F. Prelysosomal Compartments in the Unconventional Secretion of Amyloidogenic Seeds. Int J Mol Sci 2017; 18:ijms18010227. [PMID: 28124989 PMCID: PMC5297856 DOI: 10.3390/ijms18010227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/09/2017] [Accepted: 01/16/2017] [Indexed: 12/18/2022] Open
Abstract
A mechanistic link between neuron-to-neuron transmission of secreted amyloid and propagation of protein malconformation cytopathology and disease has recently been uncovered in animal models. An enormous interest in the unconventional secretion of amyloids from neurons has followed. Amphisomes and late endosomes are the penultimate maturation products of the autophagosomal and endosomal pathways, respectively, and normally fuse with lysosomes for degradation. However, under conditions of perturbed membrane trafficking and/or lysosomal deficiency, prelysosomal compartments may instead fuse with the plasma membrane to release any contained amyloid. After a brief introduction to the endosomal and autophagosomal pathways, we discuss the evidence for autophagosomal secretion (exophagy) of amyloids, with a comparative emphasis on Aβ1-42 and α-synuclein, as luminal and cytosolic amyloids, respectively. The ESCRT-mediated import of cytosolic amyloid into late endosomal exosomes, a known vehicle of transmission of macromolecules between cells, is also reviewed. Finally, mechanisms of lysosomal dysfunction, deficiency, and exocytosis are exemplified in the context of genetically identified risk factors, mainly for Parkinson's disease. Exocytosis of prelysosomal or lysosomal organelles is a last resort for clearance of cytotoxic material and alleviates cytopathy. However, they also represent a vehicle for the concentration, posttranslational modification, and secretion of amyloid seeds.
Collapse
Affiliation(s)
- Helena Borland
- Department of Neurodegeneration In Vitro, H. Lundbeck A/S, 2500 Valby, Denmark.
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, 2200N Copenhagen, Denmark.
| |
Collapse
|
136
|
Spillane KM, Tolar P. B cell antigen extraction is regulated by physical properties of antigen-presenting cells. J Cell Biol 2017; 216:217-230. [PMID: 27923880 PMCID: PMC5223605 DOI: 10.1083/jcb.201607064] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/14/2016] [Accepted: 11/17/2016] [Indexed: 01/25/2023] Open
Abstract
Antibody production and affinity maturation are driven by B cell extraction and internalization of antigen from immune synapses. However, the extraction mechanism remains poorly understood. Here we develop DNA-based nanosensors to interrogate two previously proposed mechanisms, enzymatic liberation and mechanical force. Using antigens presented by either artificial substrates or live cells, we show that B cells primarily use force-dependent extraction and resort to enzymatic liberation only if mechanical forces fail to retrieve antigen. The use of mechanical forces renders antigen extraction sensitive to the physical properties of the presenting cells. We show that follicular dendritic cells are stiff cells that promote strong B cell pulling forces and stringent affinity discrimination. In contrast, dendritic cells are soft and promote acquisition of low-affinity antigens through low forces. Thus, the mechanical properties of B cell synapses regulate antigen extraction, suggesting that distinct properties of presenting cells support different stages of B cell responses.
Collapse
MESH Headings
- Animals
- Antibody Affinity
- Antigen Presentation
- Antigens/immunology
- Antigens/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biosensing Techniques
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells, Follicular/immunology
- Dendritic Cells, Follicular/metabolism
- Elasticity
- Female
- Genotype
- Immunoglobulin kappa-Chains/genetics
- Immunoglobulin kappa-Chains/immunology
- Immunoglobulin kappa-Chains/metabolism
- Immunological Synapses/immunology
- Immunological Synapses/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nanotechnology/methods
- Phenotype
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Stress, Mechanical
- Time Factors
Collapse
Affiliation(s)
- Katelyn M Spillane
- Immune Receptor Activation Laboratory, The Francis Crick Institute, London NW1 1AT, England, UK
- Division of Immunology and Inflammation, Imperial College London, London SW7 2AZ, England, UK
| | - Pavel Tolar
- Immune Receptor Activation Laboratory, The Francis Crick Institute, London NW1 1AT, England, UK
- Division of Immunology and Inflammation, Imperial College London, London SW7 2AZ, England, UK
| |
Collapse
|
137
|
Enrich C, Rentero C, Meneses-Salas E, Tebar F, Grewal T. Annexins: Ca 2+ Effectors Determining Membrane Trafficking in the Late Endocytic Compartment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:351-385. [PMID: 29594868 DOI: 10.1007/978-3-319-55858-5_14] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite the discovery of annexins 40 years ago, we are just beginning to understand some of the functions of these still enigmatic proteins. Defined and characterized by their ability to bind anionic membrane lipids in a Ca2+-dependent manner, each annexin has to be considered a multifunctional protein, with a multitude of cellular locations and diverse activities. Underlying causes for this considerable functional diversity include their capability to associate with multiple cytosolic and membrane proteins. In recent years, the increasingly recognized establishment of membrane contact sites between subcellular compartments opens a new scenario for annexins as instrumental players to link Ca2+ signalling with the integration of membrane trafficking in many facets of cell physiology. In this chapter, we review and discuss current knowledge on the contribution of annexins in the biogenesis and functioning of the late endocytic compartment, affecting endo- and exocytic pathways in a variety of physiological consequences ranging from membrane repair, lysosomal exocytosis, to cell migration.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Elsa Meneses-Salas
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, Australia
| |
Collapse
|
138
|
Andrews NW. Detection of Lysosomal Exocytosis by Surface Exposure of Lamp1 Luminal Epitopes. Methods Mol Biol 2017; 1594:205-211. [PMID: 28456985 DOI: 10.1007/978-1-4939-6934-0_13] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elevation in the cytosolic Ca2+ concentration triggers exocytosis of lysosomes in many cell types. This chapter describes a method to detect lysosomal exocytosis in mammalian cells, which takes advantage of the presence of an abundant glycoprotein, Lamp1, on the membrane of lysosomes. Lamp1 is a transmembrane protein with a large, heavily glycosylated region that faces the lumen of lysosomes. When lysosomes fuse with the plasma membrane, epitopes present on the luminal domain of Lamp1 are exposed on the cell surface. The Lamp1 luminal epitopes can then be detected on the surface of live, unfixed cells using highly specific monoclonal antibodies and fluorescence microscopy. The main advantage of this method is its sensitivity, and the fact that it provides spatial information on lysosomal exocytosis at the single cell level.
Collapse
Affiliation(s)
- Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland at College Park, 2134 Bioscience Research Building, College Park, MD, 20742-5815, USA.
| |
Collapse
|
139
|
Soufan O, Ba-Alawi W, Afeef M, Essack M, Kalnis P, Bajic VB. DRABAL: novel method to mine large high-throughput screening assays using Bayesian active learning. J Cheminform 2016; 8:64. [PMID: 27895719 PMCID: PMC5105261 DOI: 10.1186/s13321-016-0177-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 11/03/2016] [Indexed: 11/29/2022] Open
Abstract
Background Mining high-throughput screening (HTS) assays is key for enhancing decisions in the area of drug repositioning and drug discovery. However, many challenges are encountered in the process of developing suitable and accurate methods for extracting useful information from these assays. Virtual screening and a wide variety of databases, methods and solutions proposed to-date, did not completely overcome these challenges. This study is based on a multi-label classification (MLC) technique for modeling correlations between several HTS assays, meaning that a single prediction represents a subset of assigned correlated labels instead of one label. Thus, the devised method provides an increased probability for more accurate predictions of compounds that were not tested in particular assays. Results Here we present DRABAL, a novel MLC solution that incorporates structure learning of a Bayesian network as a step to model dependency between the HTS assays. In this study, DRABAL was used to process more than 1.4 million interactions of over 400,000 compounds and analyze the existing relationships between five large HTS assays from the PubChem BioAssay Database. Compared to different MLC methods, DRABAL significantly improves the F1Score by about 22%, on average. We further illustrated usefulness and utility of DRABAL through screening FDA approved drugs and reported ones that have a high probability to interact with several targets, thus enabling drug-multi-target repositioning. Specifically DRABAL suggests the Thiabendazole drug as a common activator of the NCP1 and Rab-9A proteins, both of which are designed to identify treatment modalities for the Niemann–Pick type C disease. Conclusion We developed a novel MLC solution based on a Bayesian active learning framework to overcome the challenge of lacking fully labeled training data and exploit actual dependencies between the HTS assays. The solution is motivated by the need to model dependencies between existing experimental confirmatory HTS assays and improve prediction performance. We have pursued extensive experiments over several HTS assays and have shown the advantages of DRABAL. The datasets and programs can be downloaded from https://figshare.com/articles/DRABAL/3309562.. ![]() Electronic supplementary material The online version of this article (doi:10.1186/s13321-016-0177-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Othman Soufan
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Wail Ba-Alawi
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Moataz Afeef
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Magbubah Essack
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Panos Kalnis
- Infocloud Group, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Vladimir B Bajic
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| |
Collapse
|
140
|
Pu J, Guardia CM, Keren-Kaplan T, Bonifacino JS. Mechanisms and functions of lysosome positioning. J Cell Sci 2016; 129:4329-4339. [PMID: 27799357 DOI: 10.1242/jcs.196287] [Citation(s) in RCA: 300] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysosomes have been classically considered terminal degradative organelles, but in recent years they have been found to participate in many other cellular processes, including killing of intracellular pathogens, antigen presentation, plasma membrane repair, cell adhesion and migration, tumor invasion and metastasis, apoptotic cell death, metabolic signaling and gene regulation. In addition, lysosome dysfunction has been shown to underlie not only rare lysosome storage disorders but also more common diseases, such as cancer and neurodegeneration. The involvement of lysosomes in most of these processes is now known to depend on the ability of lysosomes to move throughout the cytoplasm. Here, we review recent findings on the mechanisms that mediate the motility and positioning of lysosomes, and the importance of lysosome dynamics for cell physiology and pathology.
Collapse
Affiliation(s)
- Jing Pu
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tal Keren-Kaplan
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
141
|
Fujiwara T, Ye S, Castro-Gomes T, Winchell CG, Andrews NW, Voth DE, Varughese KI, Mackintosh SG, Feng Y, Pavlos N, Nakamura T, Manolagas SC, Zhao H. PLEKHM1/DEF8/RAB7 complex regulates lysosome positioning and bone homeostasis. JCI Insight 2016; 1:e86330. [PMID: 27777970 DOI: 10.1172/jci.insight.86330] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mutations of the Plekhm1 gene in humans and rats cause osteopetrosis, an inherited bone disease characterized by diminished bone resorption by osteoclasts. PLEKHM1 binds to RAB7 and is critical for lysosome trafficking. However, the molecular mechanisms by which PLEKHM1 regulates lysosomal pathways remain unknown. Here, we generated germline and conditional Plekhm1-deficient mice. These mice displayed no overt abnormalities in major organs, except for an increase in trabecular bone mass. Furthermore, loss of PLEKHM1 abrogated the peripheral distribution of lysosomes and bone resorption in osteoclasts. Mechanistically, we indicated that DEF8 interacts with PLEKHM1 and promotes its binding to RAB7, whereas the binding of FAM98A and NDEL1 with PLEKHM1 connects lysosomes to microtubules. Importantly, suppression of these proteins results in lysosome positioning and bone resorption defects similar to those of Plekhm1-null osteoclasts. Thus, PLHKEM1, DEF8, FAM98A, and NDEL1 constitute a molecular complex that regulates lysosome positioning and secretion through RAB7.
Collapse
Affiliation(s)
- Toshifumi Fujiwara
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Shiqiao Ye
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | | | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | | | | | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yunfeng Feng
- Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Nathan Pavlos
- Center for Orthopedic Research, Dentistry and Health Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Takashi Nakamura
- Department of Biochemistry & Integrative Medical Biology, School of Medicine, Keio University, Tokyo, Japan
| | - Stavros C Manolagas
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Haibo Zhao
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Physiology and Biophysics, and
| |
Collapse
|
142
|
Li Y, Ozment T, Wright GL, Peterson JM. Identification of Putative Receptors for the Novel Adipokine CTRP3 Using Ligand-Receptor Capture Technology. PLoS One 2016; 11:e0164593. [PMID: 27727322 PMCID: PMC5058508 DOI: 10.1371/journal.pone.0164593] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
C1q TNF Related Protein 3 (CTRP3) is a member of a family of secreted proteins that exert a multitude of biological effects. Our initial work identified CTRP3’s promise as an effective treatment for Nonalcoholic fatty liver disease (NAFLD). Specifically, we demonstrated that mice fed a high fat diet failed to develop NAFLD when treated with CTRP3. The purpose of this current project is to identify putative receptors which mediate the hepatic actions of CTRP3.
Collapse
Affiliation(s)
- Ying Li
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, United States of America
| | - Tammy Ozment
- Quillen College of Medicine, Department of Internal Medicine, East Tennessee State University, Johnson City, Tennessee, United States of America
| | - Gary L. Wright
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, United States of America
| | - Jonathan M. Peterson
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, United States of America
- College of Public Health, Department of Health Sciences, East Tennessee State University, Johnson City, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
143
|
Boye TL, Nylandsted J. Annexins in plasma membrane repair. Biol Chem 2016; 397:961-9. [DOI: 10.1515/hsz-2016-0171] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/14/2016] [Indexed: 01/01/2023]
Abstract
Abstract
Disruption of the plasma membrane poses deadly threat to eukaryotic cells and survival requires a rapid membrane repair system. Recent evidence reveal various plasma membrane repair mechanisms, which are required for cells to cope with membrane lesions including membrane fusion and replacement strategies, remodeling of cortical actin cytoskeleton and vesicle wound patching. Members of the annexin protein family, which are Ca2+-triggered phospholipid-binding proteins emerge as important components of the plasma membrane repair system. Here, we discuss the mechanisms of plasma membrane repair involving annexins spanning from yeast to human cancer cells.
Collapse
|
144
|
Turner CT, Fuller M, Hopwood JJ, Meikle PJ, Brooks DA. Drug induced exocytosis of glycogen in Pompe disease. Biochem Biophys Res Commun 2016; 479:721-727. [PMID: 27693584 DOI: 10.1016/j.bbrc.2016.09.145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/27/2016] [Indexed: 01/11/2023]
Abstract
Pompe disease is caused by a deficiency in the lysosomal enzyme α-glucosidase, and this leads to glycogen accumulation in the autolysosomes of patient cells. Glycogen storage material is exocytosed at a basal rate in cultured Pompe cells, with one study showing up to 80% is released under specific culture conditions. Critically, exocytosis induction may reduce glycogen storage in Pompe patients, providing the basis for a therapeutic strategy whereby stored glycogen is redirected to an extracellular location and subsequently degraded by circulating amylases. The focus of the current study was to identify compounds capable of inducing rapid glycogen exocytosis in cultured Pompe cells. Here, calcimycin, lysophosphatidylcholine and α-l-iduronidase each significantly increased glycogen exocytosis compared to vehicle-treated controls. The most effective compound, calcimycin, induced exocytosis through a Ca2+-dependent mechanism, although was unable to release a pool of vesicular glycogen larger than the calcimycin-induced exocytic pore. There was reduced glycogen release from Pompe compared to unaffected cells, primarily due to increased granule size in Pompe cells. Drug induced exocytosis therefore shows promise as a therapeutic approach for Pompe patients but strategies are required to enhance the release of large molecular weight glycogen granules.
Collapse
Affiliation(s)
- Christopher T Turner
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - John J Hopwood
- Lysosomal Diseases Research Unit, SA Health and Medical Research Institute, Adelaide, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide 5001, Australia.
| |
Collapse
|
145
|
Di Venanzio G, Lazzaro M, Morales ES, Krapf D, García Véscovi E. A pore-forming toxin enables Serratia a nonlytic egress from host cells. Cell Microbiol 2016; 19. [PMID: 27532510 DOI: 10.1111/cmi.12656] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/28/2016] [Accepted: 08/12/2016] [Indexed: 01/01/2023]
Abstract
Several pathogens co-opt host intracellular compartments to survive and replicate, and they thereafter disperse progeny to prosper in a new niche. Little is known about strategies displayed by Serratia marcescens to defeat immune responses and disseminate afterwards. Upon invasion of nonphagocytic cells, Serratia multiplies within autophagosome-like vacuoles. These Serratia-containing vacuoles (SeCV) circumvent progression into acidic/degradative compartments, avoiding elimination. In this work, we show that ShlA pore-forming toxin (PFT) commands Serratia escape from invaded cells. While ShlA-dependent, Ca2+ local increase was shown in SeCVs tight proximity, intracellular Ca2+ sequestration prevented Serratia exit. Accordingly, a Ca2+ surge rescued a ShlA-deficient strain exit capacity, demonstrating that Ca2+ mobilization is essential for egress. As opposed to wild-type-SeCV, the mutant strain-vacuole was wrapped by actin filaments, showing that ShlA expression rearranges host actin. Moreover, alteration of actin polymerization hindered wild-type Serratia escape, while increased intracellular Ca2+ reorganized the mutant strain-SeCV actin distribution, restoring wild-type-SeCV phenotype. Our results demonstrate that, by ShlA expression, Serratia triggers a Ca2+ signal that reshapes cytoskeleton dynamics and ends up pushing the SeCV load out of the cell, in an exocytic-like process. These results disclose that PFTs can be engaged in allowing bacteria to exit without compromising host cell integrity.
Collapse
Affiliation(s)
| | | | - Enrique S Morales
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Darío Krapf
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Eleonora García Véscovi
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
146
|
Fan Y, He JJ. HIV-1 Tat Promotes Lysosomal Exocytosis in Astrocytes and Contributes to Astrocyte-mediated Tat Neurotoxicity. J Biol Chem 2016; 291:22830-22840. [PMID: 27609518 DOI: 10.1074/jbc.m116.731836] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/05/2016] [Indexed: 12/20/2022] Open
Abstract
Tat interaction with astrocytes has been shown to be important for Tat neurotoxicity and HIV/neuroAIDS. We have recently shown that Tat expression leads to increased glial fibrillary acidic protein (GFAP) expression and aggregation and activation of unfolded protein response/endoplasmic reticulum (ER) stress in astrocytes and causes neurotoxicity. However, the exact molecular mechanism of astrocyte-mediated Tat neurotoxicity is not defined. In this study, we showed that neurotoxic factors other than Tat protein itself were present in the supernatant of Tat-expressing astrocytes. Two-dimensional gel electrophoresis and mass spectrometry revealed significantly elevated lysosomal hydrolytic enzymes and plasma membrane-associated proteins in the supernatant of Tat-expressing astrocytes. We confirmed that Tat expression and infection of pseudotyped HIV.GFP led to increased lysosomal exocytosis from mouse astrocytes and human astrocytes. We found that Tat-induced lysosomal exocytosis was tightly coupled to astrocyte-mediated Tat neurotoxicity. In addition, we demonstrated that Tat-induced lysosomal exocytosis was astrocyte-specific and required GFAP expression and was mediated by ER stress. Taken together, these results show for the first time that Tat promotes lysosomal exocytosis in astrocytes and causes neurotoxicity through GFAP activation and ER stress induction in astrocytes and suggest a common cascade through which aberrant astrocytosis/GFAP up-regulation potentiates neurotoxicity and contributes to neurodegenerative diseases.
Collapse
Affiliation(s)
- Yan Fan
- From the Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Johnny J He
- From the Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas 76107
| |
Collapse
|
147
|
Meduri G, Guillemeau K, Dounane O, Sazdovitch V, Duyckaerts C, Chambraud B, Baulieu EE, Giustiniani J. Caspase-cleaved Tau-D(421) is colocalized with the immunophilin FKBP52 in the autophagy-endolysosomal system of Alzheimer's disease neurons. Neurobiol Aging 2016; 46:124-37. [PMID: 27479154 DOI: 10.1016/j.neurobiolaging.2016.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 04/27/2016] [Accepted: 06/24/2016] [Indexed: 01/06/2023]
Abstract
Pathologic modifications of the Tau protein leading to neurofibrillary tangle (NFT) formation are a common feature of a wide range of neurodegenerative diseases known as tauopathies, which include Alzheimer's disease (AD). We previously showed that the immunophilin FKBP52 physically and functionally interacts with Tau, and we recently reported that FKBP52 levels are abnormally low in AD patients' brains. To decipher the mechanism of FKBP52 decrease in AD brains, we performed multiple labeling immunohistofluorescence and lysosomal purification using postmortem brain samples of healthy controls (n = 8) and AD (n = 20) patients. Confocal analysis revealed that FKBP52 localizes to the endolysosomal system. We also report FKBP52 colocalization with the truncated Tau-D(421) in the autophagy-endolysosomal system in some AD neurons and that the decrease of FKBP52 correlates with NFT formation. Additional experiments of autophagy inhibition in Tau-inducible SH-SY5Y cells allowed demonstrating FKBP52 release in the extracellular milieu. Our findings point out the possibility that FKBP52 could be abnormally released from NFTs negative neurons in AD brains in correlation with the early pathologic Tau-D(421) neuronal accumulation.
Collapse
Affiliation(s)
- Geri Meduri
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Kevin Guillemeau
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Omar Dounane
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Véronique Sazdovitch
- Laboratoire de Neuropathologie Escourolle, Hôpital de La Salpêtrière, AP-HP, Paris, France
| | - Charles Duyckaerts
- Laboratoire de Neuropathologie Escourolle, Hôpital de La Salpêtrière, AP-HP, Paris, France
| | - Béatrice Chambraud
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Etienne Emile Baulieu
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France.
| | - Julien Giustiniani
- INSERM, Unité mixte de recherche 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
148
|
A rapid and sensitive assay based on particle analysis for cell degranulation detection in basophils and mast cells. Pharmacol Res 2016; 111:374-383. [PMID: 27339828 DOI: 10.1016/j.phrs.2016.05.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/24/2016] [Accepted: 05/30/2016] [Indexed: 01/02/2023]
Abstract
The degranulation of mast cells and basophils is often initiated by a number of pathophysiological responses, especially in allergic and inflammatory conditions. Efficient techniques and methods for determining the level of such degranulation are highly demanded for laboratory and clinical studies. In this work, a rapid and sensitive assay based on the particle analysis of granules in RBL-2H3 cells, a cell line widely used as a convenient model system to study the degranulation of mast cells and basophils, was developed to detect cell degranulation using a Nanosight NS300 in light scatter mode and dynamic light scattering (DLS) on a Malvern Zetasizer Nano-ZS instrument. Using this method, drug-induced mast cell degranulation and systemic anaphylaxis were efficiently determined both in cell culture medium and blood samples from animals in the current study. This promising method is expected to be widely used for screening anti-allergic and anti-inflammatory drugs both in vitro and in vivo models, as well as for determining the level of mast cell degranulation of the patients in the clinic.
Collapse
|
149
|
Miller H, Castro-Gomes T, Corrotte M, Tam C, Maugel TK, Andrews NW, Song W. Lipid raft-dependent plasma membrane repair interferes with the activation of B lymphocytes. J Cell Biol 2016; 211:1193-205. [PMID: 26694840 PMCID: PMC4687878 DOI: 10.1083/jcb.201505030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Repair of plasma membrane wounds in B lymphocytes that lack caveolin requires lysosome exocytosis and lipid raft–mediated endocytosis and inhibits activation of the B cell receptor by sequestering lipid rafts. Cells rapidly repair plasma membrane (PM) damage by a process requiring Ca2+-dependent lysosome exocytosis. Acid sphingomyelinase (ASM) released from lysosomes induces endocytosis of injured membrane through caveolae, membrane invaginations from lipid rafts. How B lymphocytes, lacking any known form of caveolin, repair membrane injury is unknown. Here we show that B lymphocytes repair PM wounds in a Ca2+-dependent manner. Wounding induces lysosome exocytosis and endocytosis of dextran and the raft-binding cholera toxin subunit B (CTB). Resealing is reduced by ASM inhibitors and ASM deficiency and enhanced or restored by extracellular exposure to sphingomyelinase. B cell activation via B cell receptors (BCRs), a process requiring lipid rafts, interferes with PM repair. Conversely, wounding inhibits BCR signaling and internalization by disrupting BCR–lipid raft coclustering and by inducing the endocytosis of raft-bound CTB separately from BCR into tubular invaginations. Thus, PM repair and B cell activation interfere with one another because of competition for lipid rafts, revealing how frequent membrane injury and repair can impair B lymphocyte–mediated immune responses.
Collapse
Affiliation(s)
- Heather Miller
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Christina Tam
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Timothy K Maugel
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| |
Collapse
|
150
|
Maass KF, Kulkarni C, Betts AM, Wittrup KD. Determination of Cellular Processing Rates for a Trastuzumab-Maytansinoid Antibody-Drug Conjugate (ADC) Highlights Key Parameters for ADC Design. AAPS J 2016; 18:635-46. [PMID: 26912181 PMCID: PMC5256610 DOI: 10.1208/s12248-016-9892-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 02/16/2016] [Indexed: 12/26/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a promising class of cancer therapeutics that combine the specificity of antibodies with the cytotoxic effects of payload drugs. A quantitative understanding of how ADCs are processed intracellularly can illustrate which processing steps most influence payload delivery, thus aiding the design of more effective ADCs. In this work, we develop a kinetic model for ADC cellular processing as well as generalizable methods based on flow cytometry and fluorescence imaging to parameterize this model. A number of key processing steps are included in the model: ADC binding to its target antigen, internalization via receptor-mediated endocytosis, proteolytic degradation of the ADC, efflux of the payload out of the cell, and payload binding to its intracellular target. The model was developed with a trastuzumab-maytansinoid ADC (TM-ADC) similar to trastuzumab-emtansine (T-DM1), which is used in the clinical treatment of HER2+ breast cancer. In three high-HER2-expressing cell lines (BT-474, NCI-N87, and SK-BR-3), we report for TM-ADC half-lives for internalization of 6-14 h, degradation of 18-25 h, and efflux rate of 44-73 h. Sensitivity analysis indicates that the internalization rate and efflux rate are key parameters for determining how much payload is delivered to a cell with TM-ADC. In addition, this model describing the cellular processing of ADCs can be incorporated into larger pharmacokinetics/pharmacodynamics models, as demonstrated in the associated companion paper.
Collapse
Affiliation(s)
- Katie F Maass
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Chethana Kulkarni
- Oncology Medicinal Chemistry, Worldwide Medicinal Chemistry, Pfizer, Groton, Connecticut, USA
| | - Alison M Betts
- Translational Research Group, Department of Pharmacokinetics Dynamics and Metabolism, Pfizer, Groton, Connecticut, USA
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave. 76-261D, Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|