51
|
Zhang W, Yu CYY, Kwok RTK, Lam JWY, Tang BZ. A photostable AIE luminogen with near infrared emission for monitoring morphological change of plasma membrane. J Mater Chem B 2018; 6:1501-1507. [DOI: 10.1039/c7tb02947k] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The morphological changes of plasma membrane can be long-term monitored by an AIEgen with high photostability.
Collapse
Affiliation(s)
- Weijie Zhang
- Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction
- Department of Chemistry
- Institute for Advanced Study and Division of Life Science
- The Hong Kong University of Science and Technology
- Clear Water Bay
| | - Chris Y. Y. Yu
- Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction
- Department of Chemistry
- Institute for Advanced Study and Division of Life Science
- The Hong Kong University of Science and Technology
- Clear Water Bay
| | - Ryan T. K. Kwok
- Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction
- Department of Chemistry
- Institute for Advanced Study and Division of Life Science
- The Hong Kong University of Science and Technology
- Clear Water Bay
| | - Jacky W. Y. Lam
- Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction
- Department of Chemistry
- Institute for Advanced Study and Division of Life Science
- The Hong Kong University of Science and Technology
- Clear Water Bay
| | - Ben Zhong Tang
- Hong Kong Branch of Chinese National Engineering Research Centre for Tissue Restoration and Reconstruction
- Department of Chemistry
- Institute for Advanced Study and Division of Life Science
- The Hong Kong University of Science and Technology
- Clear Water Bay
| |
Collapse
|
52
|
Samoylova EM, Kalsin VA, Bespalova VA, Devichensky VM, Baklaushev VP. Exosomes: from biology to clinics. GENES & CELLS 2017; 12:7-19. [DOI: 10.23868/201707024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
Exosomes are extracellular vesicles with the diameter of 30-120 nm, originating from early endosomes. Exosomes have been actively studied in the last decade, and a great amount of data has appeared on their nature and role in the intercellular transport and signaling both in the normal and pathological conditions. A particular interest to exosomes in the clinical practice emerged after the separation of their circulating fraction from the blood and the study of tumor genetic markers in them became possible (so called “liquid biopsy”). The objective of this review is to familiarize clinical specialists with the fundamentals of exosomes' biology and physiology and with the main achievements on their practical application in the medicine, as a natural drug delivery system, as well as for high-precision, early non-invasive differential diagnostics of diseases.
Collapse
|
53
|
Li Z, Zhang Y, Zhu D, Li S, Yu X, Zhao Y, Ouyang X, Xie Z, Li L. Transporting carriers for intracellular targeting delivery via non-endocytic uptake pathways. Drug Deliv 2017; 24:45-55. [PMID: 29069996 PMCID: PMC8812582 DOI: 10.1080/10717544.2017.1391889] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To develop novel therapies for clinical treatments, it increasingly depends on sophisticated delivery systems that facilitate the drugs entry into targeting cells. Profound understanding of cellular uptake routes for transporting carriers promotes the optimization of performance in drug delivery systems. Although endocytic pathway is the most important part of cellular uptake routes for many delivery systems, it suffers the trouble of enzymatic degradation of transporting carriers trapped in endosomes/lysosomes. Therefore, it is desirable to develop alternative transporting methods for delivery systems via non-endocytic pathways to achieve more effective intracellular delivery. In this review, we summarize the literature exploring transporting carriers that mediate intracellular delivery via non-endocytic pathways to present the current research status in this field. Cell-penetrating peptides, pH (low) insertion peptides, and nanoparticles are categorized to exhibit their ability to directly transport various cargos into cytoplasm via non-endocytic uptake in different cell lines. It is hoped that this review can spur the interesting on development of drug delivery systems via non-endocytic uptake pathway.
Collapse
Affiliation(s)
- Zuhong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Yanhong Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Shuiqing Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xiaopeng Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Yalei Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xiaoxi Ouyang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| |
Collapse
|
54
|
Patel D, Rorbach J, Downes K, Szukszto MJ, Pekalski ML, Minczuk M. Macropinocytic entry of isolated mitochondria in epidermal growth factor-activated human osteosarcoma cells. Sci Rep 2017; 7:12886. [PMID: 29018288 PMCID: PMC5634993 DOI: 10.1038/s41598-017-13227-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022] Open
Abstract
Mammalian mitochondria can be transferred between cells both in culture and in vivo. There is evidence that isolated mitochondria enter cells by endocytosis, but the mechanism has not been fully characterised. We investigated the entry mechanism of isolated mitochondria into human osteosarcoma (HOS) cells. Initially we confirmed that respiratory-competent cells can be produced following incubation of HOS cells lacking mitochondrial DNA (mtDNA) with functional exogenous mitochondria and selection in a restrictive medium. Treatment of HOS cells with inhibitors of different endocytic pathways suggest that uptake of EGFP-labelled mitochondria occurs via an actin-dependent endocytic pathway which is consistent with macropinocytosis. We later utilised time-lapse microscopy to show that internalised mitochondria were found in large, motile cellular vesicles. Finally, we used confocal imaging to show that EGFP-labelled mitochondria colocalise with a macropinocytic cargo molecule during internalisation, HOS cells produce membrane ruffles interacting with external mitochondria during uptake and EGFP-labelled mitochondria are found within early macropinosomes inside cells. In conclusion our results are consistent with isolated mitochondria being internalised by macropinocytosis in HOS cells.
Collapse
Affiliation(s)
- Dipali Patel
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| | - Joanna Rorbach
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Kate Downes
- CIMR, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
- Department of Haematology, University of Cambridge, NHS Blood and Transplant, Long Road, Cambridge, CB2 0PT, UK
| | - Maciej J Szukszto
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | | | - Michal Minczuk
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
55
|
Loh LN, McCarthy EMC, Narang P, Khan NA, Ward TH. Escherichia coli K1 utilizes host macropinocytic pathways for invasion of brain microvascular endothelial cells. Traffic 2017; 18:733-746. [PMID: 28799243 DOI: 10.1111/tra.12508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 01/06/2023]
Abstract
Eukaryotic cells utilize multiple endocytic pathways for specific uptake of ligands or molecules, and these pathways are commonly hijacked by pathogens to enable host cell invasion. Escherichia coli K1, a pathogenic bacterium that causes neonatal meningitis, invades the endothelium of the blood-brain barrier, but the entry route remains unclear. Here, we demonstrate that the bacteria trigger an actin-mediated uptake route, stimulating fluid phase uptake, membrane ruffling and macropinocytosis. The route of uptake requires intact lipid rafts as shown by cholesterol depletion. Using a variety of perturbants we demonstrate that small Rho GTPases and their downstream effectors have a significant effect on bacterial invasion. Furthermore, clathrin-mediated endocytosis appears to play an indirect role in E. coli K1 uptake. The data suggest that the bacteria effect a complex interplay between the Rho GTPases to increase their chances of uptake by macropinocytosis into human brain microvascular endothelial cells.
Collapse
Affiliation(s)
- Lip Nam Loh
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Elizabeth M C McCarthy
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Priyanka Narang
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Naveed A Khan
- Department of Biological Sciences, Faculty of Science and Technology, Sunway University, Selangor, Malaysia
| | - Theresa H Ward
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
56
|
Rahimi M, Regan D, Arroyo M, Subramaniam AB, Stone HA, Staykova M. Shape Transformations of Lipid Bilayers Following Rapid Cholesterol Uptake. Biophys J 2017; 111:2651-2657. [PMID: 28002741 DOI: 10.1016/j.bpj.2016.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/11/2016] [Accepted: 11/03/2016] [Indexed: 11/29/2022] Open
Abstract
High cholesterol levels in the blood increase the risk of atherosclerosis. A common explanation is that the cholesterol increase in the plasma membrane perturbs the shape and functions of cells by disrupting the cell signaling pathways and the formation of membrane rafts. In this work, we show that after enhanced transient uptake of cholesterol, mono-component lipid bilayers change their shape similarly to cell membranes in vivo. The bilayers either expel lipid protrusions or spread laterally as a result of the ensuing changes in their lipid density, the mechanical constraints imposed on them, and the properties of cyclodextrin used as a cholesterol donor. In light of the increasingly recognized link between membrane tension and cell behavior, we propose that the physical adaptation of the plasma membrane to cholesterol uptake may play a substantial role in the biological response.
Collapse
Affiliation(s)
- Mohammad Rahimi
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey
| | - David Regan
- Department of Physics, University of Durham, Durham, United Kingdom
| | - Marino Arroyo
- Universitat Politecnica de Catalunya, Barcelona, Spain
| | | | - Howard A Stone
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey
| | | |
Collapse
|
57
|
Kühn S, Lopez-Montero N, Chang YY, Sartori-Rupp A, Enninga J. Imaging macropinosomes during Shigella infections. Methods 2017; 127:12-22. [PMID: 28522322 DOI: 10.1016/j.ymeth.2017.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022] Open
Abstract
Macropinocytosis is the uptake of extracellular fluid within vesicles of varying size that takes place during numerous cellular processes in a large variety of cells. A growing number of pathogens, including viruses, parasites, and bacteria are known to induce macropinocytosis during their entry into targeted host cells. We have recently discovered that the human enteroinvasive, bacterial pathogen Shigella causes in situ macropinosome formation during its entry into epithelial cells. These infection-associated macropinosomes are not generated to ingest the bacteria, but are instead involved in Shigella's intracellular niche formation. They make contacts with the phagocytosed shigellae to promote vacuolar membrane rupture and their cytosolic release. Here, we provide an overview of the different imaging approaches that are currently used to analyze macropinocytosis during infectious processes with a focus on Shigella entry. We detail the advantages and disadvantages of genetically encoded reporters as well as chemical probes to trace fluid phase uptake. In addition, we report how such reporters can be combined with ultrastructural approaches for correlative light electron microscopy either in thin sections or within large volumes. The combined imaging techniques introduced here provide a detailed characterization of macropinosomes during bacterial entry, which, apart from Shigella, are relevant for numerous other ones, including Salmonella, Brucella or Mycobacteria.
Collapse
Affiliation(s)
- Sonja Kühn
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | | | - Yuen-Yan Chang
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Anna Sartori-Rupp
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Jost Enninga
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France.
| |
Collapse
|
58
|
Tumor Microenvironment Modulation via Gold Nanoparticles Targeting Malicious Exosomes: Implications for Cancer Diagnostics and Therapy. Int J Mol Sci 2017; 18:ijms18010162. [PMID: 28098821 PMCID: PMC5297795 DOI: 10.3390/ijms18010162] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/18/2022] Open
Abstract
Exosomes are nanovesicles formed in the endosomal pathway with an important role in paracrine and autocrine cell communication. Exosomes secreted by cancer cells, malicious exosomes, have important roles in tumor microenvironment maturation and cancer progression. The knowledge of the role of exosomes in tumorigenesis prompted a new era in cancer diagnostics and therapy, taking advantage of the use of circulating exosomes as tumor biomarkers due to their stability in body fluids and targeting malignant exosomes’ release and/or uptake to inhibit or delay tumor development. In recent years, nanotechnology has paved the way for the development of a plethora of new diagnostic and therapeutic platforms, fostering theranostics. The unique physical and chemical properties of gold nanoparticles (AuNPs) make them suitable vehicles to pursuit this goal. AuNPs’ properties such as ease of synthesis with the desired shape and size, high surface:volume ratio, and the possibility of engineering their surface as desired, potentiate AuNPs’ role in nanotheranostics, allowing the use of the same formulation for exosome detection and restraining the effect of malicious exosomes in cancer progression.
Collapse
|
59
|
Welsby I, Detienne S, N'Kuli F, Thomas S, Wouters S, Bechtold V, De Wit D, Gineste R, Reinheckel T, Elouahabi A, Courtoy PJ, Didierlaurent AM, Goriely S. Lysosome-Dependent Activation of Human Dendritic Cells by the Vaccine Adjuvant QS-21. Front Immunol 2017; 7:663. [PMID: 28105029 PMCID: PMC5215313 DOI: 10.3389/fimmu.2016.00663] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022] Open
Abstract
The adjuvant properties of the saponin QS-21 have been known for decades. It is a component of the Adjuvant System AS01 that is used in several vaccine candidates. QS-21 strongly potentiates both cellular and humoral immune responses to purified antigens, yet how it activates immune cells is largely unknown. Here, we report that QS-21 directly activated human monocyte-derived dendritic cells (moDCs) and promoted a pro-inflammatory transcriptional program. Cholesterol-dependent QS-21 endocytosis followed by lysosomal destabilization and Syk kinase activation were prerequisites for this response. Cathepsin B, a lysosomal cysteine protease, was essential for moDC activation in vitro and contributed to the adjuvant effects of QS-21 in vivo. Collectively, these findings provide new insights into the pathways involved in the direct activation of antigen-presenting cells by a clinically relevant QS-21 formulation.
Collapse
Affiliation(s)
- Iain Welsby
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) , Gosselies , Belgium
| | - Sophie Detienne
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) , Gosselies , Belgium
| | - Francisca N'Kuli
- Cell Biology Unit, de Duve Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Séverine Thomas
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) , Gosselies , Belgium
| | | | | | - Dominique De Wit
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) , Gosselies , Belgium
| | | | - Thomas Reinheckel
- Medical Faculty, Institute for Molecular Medicine and Cell Research, Albert Ludwigs University , Freiburg , Germany
| | | | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute, Université Catholique de Louvain , Brussels , Belgium
| | | | - Stanislas Goriely
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB) , Gosselies , Belgium
| |
Collapse
|
60
|
Specific uptake mechanisms of well-tolerated thermoresponsive polyglycerol-based nanogels in antigen-presenting cells of the skin. Eur J Pharm Biopharm 2016; 116:155-163. [PMID: 28027923 DOI: 10.1016/j.ejpb.2016.12.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 12/02/2016] [Accepted: 12/22/2016] [Indexed: 12/24/2022]
Abstract
Engineered nanogels are of high value for a targeted and controlled transport of compounds due to the ability to change their chemical properties by external stimuli. As it has been indicated that nanogels possess a high ability to penetrate the stratum corneum, it cannot be excluded that nanogels interact with dermal dendritic cells, especially in diseased skin. In this study the potential crosstalk of the thermoresponsive nanogels (tNGs) with the dendritic cells of the skin was investigated with the aim to determine the immunotoxicological properties of the nanogels. The investigated tNGs were made of dendritic polyglycerol (dPG) and poly(glycidyl methyl ether-co-ethyl glycidyl ether) (p(GME-co-EGE)), as polymer conferring thermoresponsive properties. Although the tNGs were taken up, they displayed neither cytotoxic and genotoxic effects nor any induction of reactive oxygen species in the tested cells. Interestingly, specific uptake mechanisms of the tNGs by the dendritic cells were depending on the nanogels cloud point temperature (Tcp), which determines the phase transition of the nanoparticle. The study points to caveolae-mediated endocytosis as being the major tNGs uptake mechanism at 37°C, which is above the Tcp of the tNGs. Remarkably, an additional uptake mechanism, beside caveolae-mediated endocytosis, was observed at 29°C, which is the Tcp of the tNGs. At this temperature, which is characterized by two different states of the tNGs, macropinocytosis was involved as well. In summary, our study highlights the impact of thermoresponsivity on the cellular uptake mechanisms which has to be taken into account if the tNGs are used as a drug delivery system.
Collapse
|
61
|
Tsujimura N, Yamada NO, Kuranaga Y, Kumazaki M, Shinohara H, Taniguchi K, Akao Y. A Novel Role of Dickkopf-Related Protein 3 in Macropinocytosis in Human Bladder Cancer T24 Cells. Int J Mol Sci 2016; 17:E1846. [PMID: 27827955 PMCID: PMC5133846 DOI: 10.3390/ijms17111846] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/22/2023] Open
Abstract
Dickkopf-related protein 3 (Dkk-3) is a potential tumor suppressor reported in various cancer entities. However, we found that Dkk-3 was exceptionally upregulated in bladder cancer T24 cells. To validate the biological role of Dkk-3 other than a tumor suppressor, we examined the function of Dkk-3 in T24 cells. Gene silencing of Dkk-3 inhibited cell growth through inducing G₀/G₁ cell-cycle arrest. Furthermore, Dkk-3 knock-down caused macropinocytosis accompanied by autophagy, which were canceled in part by their inhibitors 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and 3-methyladenine (3-MA). The macropinocytosis was induced by the Dkk-3 knock-down when there were sufficient extracellular nutrients. On the other hand, when the nutritional condition was poor, the autophagy was mainly induced by the Dkk-3 knock-down. These data indicated that Dkk-3 has a role in modulating macropinocytotic and autophagic pathways, a distinct function other than a Wnt antagonist.
Collapse
Affiliation(s)
- Nonoka Tsujimura
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Nami O Yamada
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
- Department of Anatomy, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Yuki Kuranaga
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Minami Kumazaki
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Haruka Shinohara
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Kohei Taniguchi
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu-city, Gifu 501-1194, Japan.
| |
Collapse
|
62
|
Cell Surface THY-1 Contributes to Human Cytomegalovirus Entry via a Macropinocytosis-Like Process. J Virol 2016; 90:9766-9781. [PMID: 27558416 DOI: 10.1128/jvi.01092-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/23/2016] [Indexed: 12/14/2022] Open
Abstract
Previously we showed that THY-1 has a critical role in the initial stage of infection of certain cell types with human cytomegalovirus (HCMV) and that THY-1 is important for HCMV-mediated activation of phosphatidylinositol 3-kinase (PI3K)/Akt during virus entry. THY-1 is known to interact with integrins and is a major cargo protein of clathrin-independent endocytic vesicles. Since macropinocytosis involves integrin signaling, is PI3K/Akt dependent, and is a clathrin-independent endocytic process, we determined whether THY-1 has a role in HCMV entry by macropinocytosis. Using electron microscopy in two cell lines that support HCMV infection in a THY-1-dependent manner, we found that HCMV enters these cells by a macropinocytosis-like process. THY-1 associated with HCMV virions on the cell surface and colocalized with virus inside macropinosomes. 5-(N-Ethyl-N-isopropyl)amiloride (EIPA) and soluble THY-1 blocked HCMV infection in the cell lines by ≥80% and 60%, respectively. HCMV entry into the cells triggered increased influx of extracellular fluid, a marker of macropinocytosis, and this increased fluid uptake was inhibited by EIPA and by soluble THY-1. Blocking actin depolymerization, Na+/H+ exchange, PI3K, and Pak1 kinase, which are critical for macropinocytosis, impaired HCMV infection. Neither internalized HCMV virions nor THY-1 in virus-infected cells colocalized with transferrin as determined by confocal microscopy, indicating that clathrin-mediated endocytosis was not involved in THY-1-associated virus entry. These results suggest that HCMV has adapted to utilize THY-1, a cargo protein of clathrin-independent endocytotic vesicles, to facilitate efficient entry into certain cell types by a macropinocytosis-like process. IMPORTANCE Human cytomegalovirus (HCMV) infects over half of the population and is the most common infectious cause of birth defects. The virus is the most important infection occurring in transplant recipients. The mechanism of how HCMV enters cells is controversial. In this study, we show that THY-1, a cell surface protein that is critical for the early stage of entry of HCMV into certain cell types, contributes to virus entry by macropinocytosis. Our findings suggest that HCMV has adapted to utilize THY-1 to facilitate entry of HCMV into macropinosomes in certain cell types. Further knowledge about the mechanism of HCMV entry into cells may facilitate the development of novel inhibitors of virus infection.
Collapse
|
63
|
Ha KD, Bidlingmaier SM, Liu B. Macropinocytosis Exploitation by Cancers and Cancer Therapeutics. Front Physiol 2016; 7:381. [PMID: 27672367 PMCID: PMC5018483 DOI: 10.3389/fphys.2016.00381] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022] Open
Abstract
Macropinocytosis has long been known as a primary method for cellular intake of fluid-phase and membrane-bound bulk cargo. This review seeks to re-examine the latest studies to emphasize how cancers exploit macropinocytosis to further their tumorigenesis, including details in how macropinocytosis can be adapted to serve diverse functions. Furthermore, this review will also cover the latest endeavors in targeting macropinocytosis as an avenue for novel therapeutics.
Collapse
Affiliation(s)
- Kevin D Ha
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Scott M Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Bin Liu
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
64
|
White spot syndrome virus entry is dependent on multiple endocytic routes and strongly facilitated by Cq-GABARAP in a CME-dependent manner. Sci Rep 2016; 6:28694. [PMID: 27385304 PMCID: PMC4935888 DOI: 10.1038/srep28694] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022] Open
Abstract
White spot syndrome virus (WSSV) is a lethal pathogen of shrimp and many other crustaceans, including crayfish. However, the molecular mechanism underlying its cellular entry remains elusive due to the lack of shrimp cell lines for viral propagation. Crayfish hematopoietic tissue (Hpt) cell culture was recently established as a good model for WSSV infection study. Here, we showed that multiple endocytic routes, including clathrin-mediated endocytosis (CME), macropinocytosis and caveolae-mediated endocytosis, were indispensably employed for the viral entry into Hpt cell of the crayfish Cherax quadricarinatus. Intriguingly, cellular autophagic activity was positively correlated with efficient viral entry, in which a key autophagy-related protein, γ-aminobutyric acid receptor-associated protein (Cq-GABARAP), that not only localized but also co-localized with WSSV on the Hpt cell membrane, strongly facilitated WSSV entry by binding to the viral envelope VP28 in a CME-dependent manner that was negatively regulated by Cq-Rac1. Furthermore, cytoskeletal components, including Cq-β-tubulin and Cq-β-actin, bound to both recombinant rCq-GABARAP and WSSV envelope proteins, which likely led to viral entry promotion via cooperation with rCq-GABARAP. Even under conditions that promoted viral entry, rCq-GABARAP significantly reduced viral replication at an early stage of infection, which was probably caused by the formation of WSSV aggregates in the cytoplasm.
Collapse
|
65
|
Imaging approaches for analysis of cholesterol distribution and dynamics in the plasma membrane. Chem Phys Lipids 2016; 199:106-135. [PMID: 27016337 DOI: 10.1016/j.chemphyslip.2016.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/04/2016] [Indexed: 11/21/2022]
Abstract
Cholesterol is an important lipid component of the plasma membrane (PM) of mammalian cells, where it is involved in control of many physiological processes, such as endocytosis, cell migration, cell signalling and surface ruffling. In an attempt to explain these functions of cholesterol, several models have been put forward about cholesterol's lateral and transbilayer organization in the PM. In this article, we review imaging techniques developed over the last two decades for assessing the distribution and dynamics of cholesterol in the PM of mammalian cells. Particular focus is on fluorescence techniques to study the lateral and inter-leaflet distribution of suitable cholesterol analogues in the PM of living cells. We describe also several methods for determining lateral cholesterol dynamics in the PM including fluorescence recovery after photobleaching (FRAP), fluorescence correlation spectroscopy (FCS), single particle tracking (SPT) and spot variation FCS coupled to stimulated emission depletion (STED) microscopy. For proper interpretation of such measurements, we provide some background in probe photophysics and diffusion phenomena occurring in cell membranes. In particular, we show the equivalence of the reaction-diffusion approach, as used in FRAP and FCS, and continuous time random walk (CTRW) models, as often invoked in SPT studies. We also discuss mass spectrometry (MS) based imaging of cholesterol in the PM of fixed cells and compare this method with fluorescence imaging of sterols. We conclude that evidence from many experimental techniques converges towards a model of a homogeneous distribution of cholesterol with largely free and unhindered diffusion in both leaflets of the PM.
Collapse
|
66
|
Lange Y, Steck TL. Active membrane cholesterol as a physiological effector. Chem Phys Lipids 2016; 199:74-93. [PMID: 26874289 DOI: 10.1016/j.chemphyslip.2016.02.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 02/05/2023]
Abstract
Sterols associate preferentially with plasma membrane sphingolipids and saturated phospholipids to form stoichiometric complexes. Cholesterol in molar excess of the capacity of these polar bilayer lipids has a high accessibility and fugacity; we call this fraction active cholesterol. This review first considers how active cholesterol serves as an upstream regulator of cellular sterol homeostasis. The mechanism appears to utilize the redistribution of active cholesterol down its diffusional gradient to the endoplasmic reticulum and mitochondria, where it binds multiple effectors and directs their feedback activity. We have also reviewed a broad literature in search of a role for active cholesterol (as opposed to bulk cholesterol or lipid domains such as rafts) in the activity of diverse membrane proteins. Several systems provide such evidence, implicating, in particular, caveolin-1, various kinds of ABC-type cholesterol transporters, solute transporters, receptors and ion channels. We suggest that this larger role for active cholesterol warrants close attention and can be tested easily.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, 1653 W. Congress Parkway, Chicago, IL 60612, USA.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
67
|
Rosazza C, Deschout H, Buntz A, Braeckmans K, Rols MP, Zumbusch A. Endocytosis and Endosomal Trafficking of DNA After Gene Electrotransfer In Vitro. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e286. [PMID: 26859199 PMCID: PMC4884790 DOI: 10.1038/mtna.2015.59] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 01/08/2023]
Abstract
DNA electrotransfer is a successful technique for gene delivery into cells and represents an attractive alternative to virus-based methods for clinical applications including gene therapy and DNA vaccination. However, little is currently known about the mechanisms governing DNA internalization and its fate inside cells. The objectives of this work were to investigate the role of endocytosis and to quantify the contribution of different routes of cellular trafficking during DNA electrotransfer. To pursue these objectives, we performed flow cytometry and single-particle fluorescence microscopy experiments using inhibitors of endocytosis and endosomal markers. Our results show that ~50% of DNA is internalized by caveolin/raft-mediated endocytosis, 25% by clathrin-mediated endocytosis, and 25% by macropinocytosis. During active transport, DNA is routed through multiple endosomal compartments with, in the hour following electrotransfer, 70% found in Rab5 structures, 50% in Rab11-containing organelles and 30% in Rab9 compartments. Later, 60% of DNA colocalizes with Lamp1 vesicles. Because these molecular markers can overlap while following organelles through several steps of trafficking, the percentages do not sum up to 100%. We conclude that electrotransferred DNA uses the classical endosomal trafficking pathways. Our results are important for a generalized understanding of gene electrotransfer, which is crucial for its safe use in clinics.
Collapse
Affiliation(s)
- Christelle Rosazza
- Department of Chemistry, University of Konstanz, Konstanz, Germany.,Department of Structural Biology and Biophysics, Institute of Pharmacology and Structural Biology (IPBS), CNRS UMR5089, Toulouse, France.,University of Toulouse III, UPS, Toulouse, France
| | - Hendrik Deschout
- Laboratory of General Biochemistry and Physical Pharmacy, Department of Pharmaceutics, University of Ghent, Ghent, Belgium
| | - Annette Buntz
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Department of Pharmaceutics, University of Ghent, Ghent, Belgium
| | - Marie-Pierre Rols
- Department of Structural Biology and Biophysics, Institute of Pharmacology and Structural Biology (IPBS), CNRS UMR5089, Toulouse, France.,University of Toulouse III, UPS, Toulouse, France
| | - Andreas Zumbusch
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| |
Collapse
|
68
|
Han SC, Guo HC, Sun SQ, Jin Y, Wei YQ, Feng X, Yao XP, Cao SZ, Xiang Liu D, Liu XT. Productive Entry of Foot-and-Mouth Disease Virus via Macropinocytosis Independent of Phosphatidylinositol 3-Kinase. Sci Rep 2016; 6:19294. [PMID: 26757826 PMCID: PMC4725844 DOI: 10.1038/srep19294] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/08/2015] [Indexed: 12/12/2022] Open
Abstract
Virus entry is an attractive target for therapeutic intervention. Here, using a combination of electron microscopy, immunofluorescence assay, siRNA interference, specific pharmacological inhibitors, and dominant negative mutation, we demonstrated that the entry of foot-and-mouth disease virus (FMDV) triggered a substantial amount of plasma membrane ruffling. We also found that the internalization of FMDV induced a robust increase in fluid-phase uptake, and virions internalized within macropinosomes colocalized with phase uptake marker dextran. During this stage, the Rac1-Pak1 signaling pathway was activated. After specific inhibition on actin, Na(+)/H(+) exchanger, receptor tyrosine kinase, Rac1, Pak1, myosin II, and protein kinase C, the entry and infection of FMDV significantly decreased. However, inhibition of phosphatidylinositol 3-kinase (PI3K) did not reduce FMDV internalization but increased the viral entry and infection to a certain extent, implying that FMDV entry did not require PI3K activity. Results showed that internalization of FMDV exhibited the main hallmarks of macropinocytosis. Moreover, intracellular trafficking of FMDV involves EEA1/Rab5-positive vesicles. The present study demonstrated macropinocytosis as another endocytic pathway apart from the clathrin-mediated pathway. The findings greatly expand our understanding of the molecular mechanisms of FMDV entry into cells, as well as provide potential insights into the entry mechanisms of other picornaviruses.
Collapse
Affiliation(s)
- Shi-Chong Han
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hui-Chen Guo
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shi-Qi Sun
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ye Jin
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yan-Quan Wei
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xia Feng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xue-Ping Yao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Sui-Zhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Ding Xiang Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Xiang-Tao Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
69
|
Carquin M, D'Auria L, Pollet H, Bongarzone ER, Tyteca D. Recent progress on lipid lateral heterogeneity in plasma membranes: From rafts to submicrometric domains. Prog Lipid Res 2015; 62:1-24. [PMID: 26738447 DOI: 10.1016/j.plipres.2015.12.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/29/2022]
Abstract
The concept of transient nanometric domains known as lipid rafts has brought interest to reassess the validity of the Singer-Nicolson model of a fluid bilayer for cell membranes. However, this new view is still insufficient to explain the cellular control of surface lipid diversity or membrane deformability. During the past decades, the hypothesis that some lipids form large (submicrometric/mesoscale vs nanometric rafts) and stable (>min vs s) membrane domains has emerged, largely based on indirect methods. Morphological evidence for stable submicrometric lipid domains, well-accepted for artificial and highly specialized biological membranes, was further reported for a variety of living cells from prokaryot es to yeast and mammalian cells. However, results remained questioned based on limitations of available fluorescent tools, use of poor lipid fixatives, and imaging artifacts due to non-resolved membrane projections. In this review, we will discuss recent evidence generated using powerful and innovative approaches such as lipid-specific toxin fragments that support the existence of submicrometric domains. We will integrate documented mechanisms involved in the formation and maintenance of these domains, and provide a perspective on their relevance on membrane deformability and regulation of membrane protein distribution.
Collapse
Affiliation(s)
- Mélanie Carquin
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ludovic D'Auria
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Hélène Pollet
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Ernesto R Bongarzone
- The Myelin Regeneration Group at the Dept. Anatomy & Cell Biology, College of Medicine, University of Illinois, 808 S. Wood St. MC512, Chicago, IL. 60612. USA
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| |
Collapse
|
70
|
Martin TM, Plautz SA, Pannier AK. Temporal endogenous gene expression profiles in response to lipid-mediated transfection. J Gene Med 2015; 17:14-32. [PMID: 25663588 DOI: 10.1002/jgm.2821] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Design of efficient nonviral gene delivery systems is limited as a result of the rudimentary understanding of the specific molecules and processes that facilitate DNA transfer. METHODS Lipoplexes formed with Lipofectamine 2000 (LF2000) and plasmid-encoding green fluorescent protein (GFP) were delivered to the HEK 293T cell line. After treating cells with lipoplexes, HG-U133 Affymetrix microarrays were used to identify endogenous genes differentially expressed between treated and untreated cells (2 h exposure) or between flow-separated transfected cells (GFP+) and treated, untransfected cells (GFP-) at 8, 16 and 24 h after lipoplex treatment. Cell priming studies were conducted using pharmacologic agents to alter endogenous levels of the identified differentially expressed genes to determine effect on transfection levels. RESULTS Relative to untreated cells 2 h after lipoplex treatment, only downregulated genes were identified ≥ 30-fold: ALMS1, ITGB1, FCGR3A, DOCK10 and ZDDHC13. Subsequently, relative to GFP- cells, the GFP+ cell population showed at least a five-fold upregulation of RAP1A and PACSIN3 (8 h) or HSPA6 and RAP1A (16 and 24 h). Pharmacologic studies altering endogenous levels for ALMS1, FCGR3A, and DOCK10 (involved in filopodia protrusions), ITGB1 (integrin signaling), ZDDHC13 (membrane trafficking) and PACSIN3 (proteolytic shedding of membrane receptors) were able to increase or decrease transgene production. CONCLUSIONS RAP1A, PACSIN3 and HSPA6 may help lipoplex-treated cells overcome a transcriptional shutdown due to treatment with lipoplexes and provide new targets for investigating molecular mechanisms of transfection or for enhancing transfection through cell priming or engineering of the nonviral gene delivery system.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Pharmaceutical Sciences, Durham Research Center II, University of Nebraska-Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
71
|
Martin TM, Plautz SA, Pannier AK. Temporal endogenous gene expression profiles in response to polymer-mediated transfection and profile comparison to lipid-mediated transfection. J Gene Med 2015; 17:33-53. [PMID: 25663627 DOI: 10.1002/jgm.2822] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Design of efficient nonviral gene delivery systems is limited by the rudimentary understanding of specific molecules that facilitate transfection. METHODS Polyplexes using 25-kDa polyethylenimine (PEI) and plasmid-encoding green fluorescent protein (GFP) were delivered to HEK 293T cells. After treating cells with polyplexes, microarrays were used to identify endogenous genes differentially expressed between treated and untreated cells (2 h of exposure) or between flow-separated transfected cells (GFP+) and treated, untransfected cells (GFP-) at 8, 16 and 24 h after lipoplex treatment. Cell priming studies were conducted using pharmacologic agents to alter endogenous levels of the identified differentially expressed genes to determine effect on transfection levels. Differentially expressed genes in polyplex-mediated transfection were compared with those differentially expressed in lipoplex transfection to identify DNA carrier-dependent molecular factors. RESULTS Differentially expressed genes were RGS1, ARHGAP24, PDZD2, SNX24, GSN and IGF2BP1 after 2 h; RAP1A and ACTA1 after 8 h; RAP1A, WDR78 and ACTA1 after 16 h; and RAP1A, SCG5, ATF3, IREB2 and ACTA1 after 24 h. Pharmacologic studies altering endogenous levels for ARHGAP24, GSN, IGF2BP1, PDZD2 and RGS1 were able to increase or decrease transgene production. Comparing differentially expressed genes for polyplexes and lipoplexes, no common genes were identified at the 2-h time point, whereas, after the 8-h time point, RAP1A, ATF3 and HSPA6 were similarly expressed. SCG5 and PGAP1 were only upregulated in polyplex-transfected cells. CONCLUSIONS The identified genes and pharmacologic agents provide targets for improving transfection systems, although polyplex or lipoplex dependencies must be considered.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Pharmaceutical Sciences, Durham Research Center II, University of Nebraska-Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
72
|
Mazuryk O, Suzenet F, Kieda C, Brindell M. The biological effect of the nitroimidazole derivative of a polypyridyl ruthenium complex on cancer and endothelial cells. Metallomics 2015; 7:553-66. [PMID: 25711770 DOI: 10.1039/c5mt00037h] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The ruthenium polypyridyl complexes [Ru(dip)2(bpy/bpy-2-nitroIm)](2+) (dip = 4,7-diphenyl-1,10-phenanthroline, bpy = 2,2'-bipyridine, bpy-2-nitroIm = 4-[3-(2-nitro-1H-imidazol-1-yl)propyl]) were found to be ca. ten times more cytotoxic against breast cancer (4T1) and human lung adenocarcinoma epithelial cells (A549) than a well-known anticancer drug, cisplatin. Even though the Ru complexes were quite cytotoxic towards FVB mouse lung microvascular endothelial cells (MLuMEC FVB) their efflux from these non transformed cells was much more efficient than from cancer ones. Both Ru complexes accumulated in cells. The cellular uptake of both Ru complexes occurs through passive diffusion while the nitroimidazole derivative is also endocytosed. They arrest cell growth in the S-phase and induce apoptosis. Such cell response can result from activation of oxidative stress by Ru complexes. The modulation of the mRNA expression profile for genes which might be involved in metastasis and angiogenesis processes by Ru complexes was analyzed for both cancer (4T1) and endothelial (MLuMEC FVB) cells. Ru complexes appeared to have a distinct impact on cell adhesion and migration as well as they affect endothelial cell vasculature. They are not only cytotoxic but are also potentially invasive and anti-metastatic agents. This work illustrates the putative future development of polypyridyl ruthenium.
Collapse
Affiliation(s)
- Olga Mazuryk
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Krakow, Poland.
| | | | | | | |
Collapse
|
73
|
Carquin M, Conrard L, Pollet H, Van Der Smissen P, Cominelli A, Veiga-da-Cunha M, Courtoy PJ, Tyteca D. Cholesterol segregates into submicrometric domains at the living erythrocyte membrane: evidence and regulation. Cell Mol Life Sci 2015; 72:4633-51. [PMID: 26077601 PMCID: PMC11113096 DOI: 10.1007/s00018-015-1951-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/07/2015] [Accepted: 06/04/2015] [Indexed: 01/01/2023]
Abstract
Although cholesterol is essential for membrane fluidity and deformability, the level of its lateral heterogeneity at the plasma membrane of living cells is poorly understood due to lack of appropriate probe. We here report on the usefulness of the D4 fragment of Clostridium perfringens toxin fused to mCherry (theta*), as specific, non-toxic, sensitive and quantitative cholesterol-labeling tool, using erythrocyte flat membrane. By confocal microscopy, theta* labels cholesterol-enriched submicrometric domains in coverslip-spread but also gel-suspended (non-stretched) fresh erythrocytes, suggesting in vivo relevance. Cholesterol domains on spread erythrocytes are stable in time and space, restricted by membrane:spectrin anchorage via 4.1R complexes, and depend on temperature and sphingomyelin, indicating combined regulation by extrinsic membrane:cytoskeleton interaction and by intrinsic lipid packing. Cholesterol domains partially co-localize with BODIPY-sphingomyelin-enriched domains. In conclusion, we show that theta* is a useful vital probe to study cholesterol organization and demonstrate that cholesterol forms submicrometric domains in living cells.
Collapse
Affiliation(s)
- Mélanie Carquin
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Louise Conrard
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Hélène Pollet
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Patrick Van Der Smissen
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Antoine Cominelli
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Maria Veiga-da-Cunha
- Laboratory of Physiological Chemistry, de Duve Institute and Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Pierre J Courtoy
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute and Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, 1200, Brussels, Belgium.
| |
Collapse
|
74
|
Kettle E, Page SL, Morgan GP, Malladi CS, Wong CL, Boadle RA, Marsh BJ, Robinson PJ, Chircop M. A Cholesterol-Dependent Endocytic Mechanism Generates Midbody Tubules During Cytokinesis. Traffic 2015; 16:1174-92. [DOI: 10.1111/tra.12328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/24/2015] [Accepted: 08/24/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Emma Kettle
- Children's Medical Research Institute; The University of Sydney; 214 Hawkesbury Road Westmead NSW 2145 Australia
| | - Scott L. Page
- Children's Medical Research Institute; The University of Sydney; 214 Hawkesbury Road Westmead NSW 2145 Australia
| | - Garry P. Morgan
- Institute for Molecular Biosciences, Queensland Bioscience Precinct; The University of Queensland; Brisbane Queensland 4072 Australia
| | - Chandra S. Malladi
- Department of Molecular Physiology, School of Medicine; University of Western Sydney; Penrith NSW 2751 Australia
| | - Chin L. Wong
- Children's Medical Research Institute; The University of Sydney; 214 Hawkesbury Road Westmead NSW 2145 Australia
| | - Ross A. Boadle
- Westmead Millennium Institute for Medical Research; 176 Hawkesbury Road Westmead NSW 2145 Australia
| | - Brad J. Marsh
- Institute for Molecular Biosciences, Queensland Bioscience Precinct; The University of Queensland; Brisbane Queensland 4072 Australia
| | - Phillip J. Robinson
- Children's Medical Research Institute; The University of Sydney; 214 Hawkesbury Road Westmead NSW 2145 Australia
| | - Megan Chircop
- Children's Medical Research Institute; The University of Sydney; 214 Hawkesbury Road Westmead NSW 2145 Australia
| |
Collapse
|
75
|
Rewatkar PV, Parton RG, Parekh HS, Parat MO. Are caveolae a cellular entry route for non-viral therapeutic delivery systems? Adv Drug Deliv Rev 2015; 91:92-108. [PMID: 25579057 DOI: 10.1016/j.addr.2015.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/23/2014] [Accepted: 01/02/2015] [Indexed: 12/20/2022]
Abstract
The development of novel therapies increasingly relies on sophisticated delivery systems that allow the drug or gene expression-modifying agent of interest entry into cells. These systems can promote cellular targeting and/or entry, and they vary in size, charge, and functional group chemistry. Their optimization requires an in depth knowledge of the cellular routes of entry in normal and pathological states. Caveolae are plasma membrane invaginations that have the potential to undergo endocytosis. We critically review the literature exploring whether drug or nucleic acid delivery systems exploit and/or promote cellular entry via caveolae. A vast majority of studies employ pharmacological tools, co-localization experiments and very few make use of molecular tools. We provide clarification on how results of such studies should be interpreted and make suggestions for future studies.
Collapse
Affiliation(s)
- Prarthana V Rewatkar
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia
| | - Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, QLD 4072 Australia.
| | - Harendra S Parekh
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia.
| | - Marie-Odile Parat
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
76
|
Ercole F, Whittaker MR, Quinn JF, Davis TP. Cholesterol Modified Self-Assemblies and Their Application to Nanomedicine. Biomacromolecules 2015; 16:1886-914. [DOI: 10.1021/acs.biomac.5b00550] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Francesca Ercole
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Michael R. Whittaker
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - John F. Quinn
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas P. Davis
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department
of Chemistry, University of Warwick, Coventry, ULCV4 7AL, United Kingdom
| |
Collapse
|
77
|
Han J, Shuvaev VV, Davies PF, Eckmann DM, Muro S, Muzykantov VR. Flow shear stress differentially regulates endothelial uptake of nanocarriers targeted to distinct epitopes of PECAM-1. J Control Release 2015; 210:39-47. [PMID: 25966362 DOI: 10.1016/j.jconrel.2015.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 01/01/2023]
Abstract
Targeting nanocarriers (NC) to endothelial cell adhesion molecules including Platelet-Endothelial Cell Adhesion Molecule-1 (PECAM-1 or CD31) improves drug delivery and pharmacotherapy of inflammation, oxidative stress, thrombosis and ischemia in animal models. Recent studies unveiled that hydrodynamic conditions modulate endothelial endocytosis of NC targeted to PECAM-1, but the specificity and mechanism of effects of flow remain unknown. Here we studied the effect of flow on endocytosis by human endothelial cells of NC targeted by monoclonal antibodies Ab62 and Ab37 to distinct epitopes on the distal extracellular domain of PECAM. Flow in the range of 1-8dyn/cm(2), typical for venous vasculature, stimulated the uptake of spherical Ab/NC (~180nm diameter) carrying ~50 vs 200 Ab62 and Ab37 per NC, respectively. Effect of flow was inhibited by disruption of cholesterol-rich plasmalemma domains and deletion of PECAM-1 cytosolic tail. Flow stimulated endocytosis of Ab62/NC and Ab37/NC via eliciting distinct signaling pathways mediated by RhoA/ROCK and Src Family Kinases, respectively. Therefore, flow stimulates endothelial endocytosis of Ab/NC in a PECAM-1 epitope specific manner. Using ligands of binding to distinct epitopes on the same target molecule may enable fine-tuning of intracellular delivery based on the hemodynamic conditions in the vascular area of interest.
Collapse
Affiliation(s)
- Jingyan Han
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA19104, USA; Vascular Biology Section, Department of Medicine, Boston University, Boston, MA 02421, USA
| | - Vladimir V Shuvaev
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA19104, USA
| | - Peter F Davies
- Department of Pathology & Lab Medicine and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA19104, USA
| | - David M Eckmann
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, USA
| | - Silvia Muro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Vladimir R Muzykantov
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA19104, USA.
| |
Collapse
|
78
|
Yue HY, Xu J. Cholesterol regulates multiple forms of vesicle endocytosis at a mammalian central synapse. J Neurochem 2015; 134:247-60. [PMID: 25893258 DOI: 10.1111/jnc.13129] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/01/2015] [Accepted: 03/31/2015] [Indexed: 01/10/2023]
Abstract
Endocytosis in synapses sustains neurotransmission by recycling vesicle membrane and maintaining the homeostasis of synaptic membrane. A role of membrane cholesterol in synaptic endocytosis remains controversial because of conflicting observations, technical limitations in previous studies, and potential interference from non-specific effects after cholesterol manipulation. Furthermore, it remains unclear whether cholesterol participates in distinct forms of endocytosis that function under different activity levels. In this study, applying the whole-cell membrane capacitance measurement to monitor endocytosis in real time at the rat calyx of Held terminals, we found that disrupting cholesterol with dialysis of cholesterol oxidase or methyl-β-cyclodextrin impaired three different forms of endocytosis, including slow endocytosis, rapid endocytosis, and endocytosis of the retrievable membrane that exists at the surface before stimulation. The effects were observed when disruption of cholesterol was mild enough not to change Ca(2+) channel current or vesicle exocytosis, indicative of stringent cholesterol requirement in synaptic endocytosis. Extracting cholesterol with high concentrations of methyl-β-cyclodextrin reduced exocytosis, mainly by decreasing the readily releasable pool and the vesicle replenishment after readily releasable pool depletion. Our study suggests that cholesterol is an important, universal regulator in multiple forms of vesicle endocytosis at mammalian central synapses.
Collapse
Affiliation(s)
- Hai-Yuan Yue
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Jianhua Xu
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia, USA.,Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
79
|
Rastew E, Morf L, Singh U. Entamoeba histolytica rhomboid protease 1 has a role in migration and motility as validated by two independent genetic approaches. Exp Parasitol 2015; 154:33-42. [PMID: 25889553 DOI: 10.1016/j.exppara.2015.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/24/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022]
Abstract
Rhomboid proteins represent a recently discovered family of intramembrane proteases present in a broad range of organisms and with increasing links to human diseases. The enteric parasite Entamoeba histolytica has evolved multiple mechanisms to adapt to the human host environment and establish infection. Our recent studies identified EhROM1 as a functional E. histolytica rhomboid protease with roles in adhesion to and phagocytosis of host cells. Since those studies were performed in a non-virulent strain, roles in parasite virulence could not be assessed. We focused this study on the comparison and validation of two genetic manipulation techniques: overexpression of a dominant-negative catalytic mutant of EhROM1 and knock down of EhROM1 using a RNAi-based silencing approach followed by functional studies of phenotypic analyses in virulent parasites. Both the EhROM1 catalytic mutant and parasites with EhROM1 downregulation were reduced in cytotoxicity, hemolytic activity, and directional and non-directional transwell migration. Importantly, the role for EhROM1 in cell migration mimics similar roles for rhomboid proteases from mammalian and apicomplexan systems. However, the EhROM1 catalytic mutant and EhROM1 downregulation parasites had different phenotypes for erythrophagocytosis, while complement resistance was not affected in either strain. In summary, in this study we genetically manipulated E. histolytica rhomboid protease EhROM1 by two different approaches and identified similarly attenuated phenotypes by both approaches, suggesting a novel role for EhROM1 in amebic motility.
Collapse
Affiliation(s)
- Elena Rastew
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Morf
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Upinder Singh
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
80
|
Preta G, Cronin JG, Sheldon IM. Dynasore - not just a dynamin inhibitor. Cell Commun Signal 2015; 13:24. [PMID: 25889964 PMCID: PMC4396812 DOI: 10.1186/s12964-015-0102-1] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/26/2015] [Indexed: 12/23/2022] Open
Abstract
Dynamin is a GTPase protein that is essential for membrane fission during clathrin-mediated endocytosis in eukaryotic cells. Dynasore is a GTPase inhibitor that rapidly and reversibly inhibits dynamin activity, which prevents endocytosis. However, comparison between cells treated with dynasore and RNA interference of genes encoding dynamin, reveals evidence that dynasore reduces labile cholesterol in the plasma membrane, and disrupts lipid raft organization, in a dynamin-independent manner. To explore the role of dynamin it is important to use multiple dynamin inhibitors, alongside the use of dynamin mutants and RNA interference targeting genes encoding dynamin. On the other hand, dynasore provides an interesting tool to explore the regulation of cholesterol in plasma membranes.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| | - James G Cronin
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| | - I Martin Sheldon
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| |
Collapse
|
81
|
de Carvalho TMU, Barrias ES, de Souza W. Macropinocytosis: a pathway to protozoan infection. Front Physiol 2015; 6:106. [PMID: 25914647 PMCID: PMC4391238 DOI: 10.3389/fphys.2015.00106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/17/2015] [Indexed: 11/13/2022] Open
Abstract
Among the various endocytic mechanisms in mammalian cells, macropinocytosis involves internalization of large amounts of plasma membrane together with extracellular medium, leading to macropinosome formation. These structures are formed when plasma membrane ruffles are assembled after actin filament rearrangement. In dendritic cells, macropinocytosis has been reported to play a role in antigen presentation. Several intracellular pathogens are internalized by host cells via multiple endocytic pathways and macropinocytosis has been described as an important entry site for various organisms. Some bacteria, such as Legionella pneumophila, as well as various viruses, use this pathway to penetrate and subvert host cells. Some protozoa, which are larger than bacteria and virus, can also use this pathway to invade host cells. As macropinocytosis is characterized by the formation of large uncoated vacuoles and is triggered by various signaling pathways, which is similar to what occurs during the formation of the majority of parasitophorous vacuoles, it is believed that this phenomenon may be more widely used by parasites than is currently appreciated. Here we review protozoa host cell invasion via macropinocytosis.
Collapse
Affiliation(s)
- Tecia M U de Carvalho
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciência da Saúde, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil ; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens e Centro Nacional de Bioimagens-CENABIO, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Emile S Barrias
- Instituto Nacional de Metrologia, Qualidade e Tecnologia-Inmetro Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciência da Saúde, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil ; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens e Centro Nacional de Bioimagens-CENABIO, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
82
|
Shubin AV, Demidyuk IV, Lunina NA, Komissarov AA, Roschina MP, Leonova OG, Kostrov SV. Protease 3C of hepatitis A virus induces vacuolization of lysosomal/endosomal organelles and caspase-independent cell death. BMC Cell Biol 2015; 16:4. [PMID: 25886889 PMCID: PMC4355371 DOI: 10.1186/s12860-015-0050-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 01/26/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND 3C proteases, the main proteases of picornaviruses, play the key role in viral life cycle by processing polyproteins. In addition, 3C proteases digest certain host cell proteins to suppress antiviral defense, transcription, and translation. The activity of 3C proteases per se induces host cell death, which makes them critical factors of viral cytotoxicity. To date, cytotoxic effects have been studied for several 3C proteases, all of which induce apoptosis. This study for the first time describes the cytotoxic effect of 3C protease of human hepatitis A virus (3Cpro), the only proteolytic enzyme of the virus. RESULTS Individual expression of 3Cpro induced catalytic activity-dependent cell death, which was not abrogated by the pan-caspase inhibitor (z-VAD-fmk) and was not accompanied by phosphatidylserine externalization in contrast to other picornaviral 3C proteases. The cell survival was also not affected by the inhibitors of cysteine proteases (z-FA-fmk) and RIP1 kinase (necrostatin-1), critical enzymes involved in non-apoptotic cell death. A substantial fraction of dying cells demonstrated numerous non-acidic cytoplasmic vacuoles with not previously described features and originating from several types of endosomal/lysosomal organelles. The lysosomal protein Lamp1 and GTPases Rab5, Rab7, Rab9, and Rab11 were associated with the vacuolar membranes. The vacuolization was completely blocked by the vacuolar ATPase inhibitor (bafilomycin A1) and did not depend on the activity of the principal factors of endosomal transport, GTPases Rab5 and Rab7, as well as on autophagy and macropinocytosis. CONCLUSIONS 3Cpro, apart from other picornaviral 3C proteases, induces caspase-independent cell death, accompanying by cytoplasmic vacuolization. 3Cpro-induced vacuoles have unique properties and are formed from several organelle types of the endosomal/lysosomal compartment. The data obtained demonstrate previously undocumented morphological characters of the 3Cpro-induced cell death, which can reflect unknown aspects of the human hepatitis A virus-host cell interaction.
Collapse
Affiliation(s)
- Andrey V Shubin
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Ilya V Demidyuk
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Nataliya A Lunina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Alexey A Komissarov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Marina P Roschina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Olga G Leonova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119992, Russia.
| | - Sergey V Kostrov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
- National Research Center "Kurchatov Institute", Moscow, 123182, Russia.
| |
Collapse
|
83
|
Abstract
There are currently no nanoparticle formulations that optimally target diseased cells in the body. A small percentage of nanoparticles reach these cells and most accumulate in cells of the mononuclear phagocytic system. This chapter explores the interactions between nanoparticles and cells that may explain the causes for off-target accumulation of nanoparticles. A greater understanding of the nanoparticle-cellular interactions will lead to improvements in particle design for improved therapeutic outcome.
Collapse
Affiliation(s)
- Abdullah Syed
- Institute of Biomaterials and Biomedical Engineering, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College St., 407, Toronto, ON, M5S 3G9, Canada
| | | |
Collapse
|
84
|
Egami Y, Taguchi T, Maekawa M, Arai H, Araki N. Small GTPases and phosphoinositides in the regulatory mechanisms of macropinosome formation and maturation. Front Physiol 2014; 5:374. [PMID: 25324782 PMCID: PMC4179697 DOI: 10.3389/fphys.2014.00374] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/10/2014] [Indexed: 12/26/2022] Open
Abstract
Macropinosome formation requires the sequential activation of numerous signaling pathways that coordinate the actin-driven formation of plasma membrane protrusions (ruffles) and circular ruffles (macropinocytic cups), followed by the closure of these macropinocytic cups into macropinosomes. In the process of macropinosome formation, localized productions of phosphoinositides such as PI(4,5)P2 and PI(3,4,5)P3 spatiotemporally orchestrate actin polymerization and rearrangement through recruiting and activating a variety of actin-associated proteins. In addition, the sequential activation of small GTPases, which are known to be master regulators of the actin cytoskeleton, plays a pivotal role in parallel with phosphoinositides. To complete macropinosome formation, phosphoinositide breakdown and Rho GTPase deactivation must occur in appropriate timings. After the nascent macropinosomes are formed, phosphoinositides and several Rab GTPases control macropinosome maturation by regulating vesicle trafficking and membrane fusion. In this review, we summarize recent advances in our understanding of the critical functions of phosphoinositide metabolism and small GTPases in association with their downstream effectors in macropinocytosis.
Collapse
Affiliation(s)
- Youhei Egami
- Department of Histology and Cell Biology, School of Medicine, Kagawa University Miki, Japan
| | - Tomohiko Taguchi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo Tokyo, Japan ; Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, University of Tokyo Tokyo, Japan
| | - Masashi Maekawa
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo Tokyo, Japan ; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital Toronto, ON, Canada
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo Tokyo, Japan ; Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, University of Tokyo Tokyo, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University Miki, Japan
| |
Collapse
|
85
|
Entry of Bombyx mori nucleopolyhedrovirus into BmN cells by cholesterol-dependent macropinocytic endocytosis. Biochem Biophys Res Commun 2014; 453:166-71. [PMID: 25264104 DOI: 10.1016/j.bbrc.2014.09.073] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/18/2014] [Indexed: 11/19/2022]
Abstract
Bombyx mori nucleopolyhedrovirus (BmNPV) is a serious viral pathogen of silkworm, and no drug or specific protection against BmNPV infection is available at present time. Although functions of most BmNPV genes were depicted in recent years, knowledge on the mechanism of BmNPV entry into insect cells is still limited. Here BmNPV cell entry mechanism is investigated by different endocytic inhibitor application and subcellular analysis. Results indicated that BmNPV enters BmN cells by clathrin-independent macropinocytic endocytosis, which is mediated by cholesterol in a dose-dependent manner, and cholesterol replenishment rescued the BmNPV infection partially.
Collapse
|
86
|
Zhang X, Wang C, Jin L, Han Z, Xiao Y. Photostable bipolar fluorescent probe for video tracking plasma membranes related cellular processes. ACS APPLIED MATERIALS & INTERFACES 2014; 6:12372-12379. [PMID: 25039476 DOI: 10.1021/am503849c] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Plasma membranes can sense the stimulations and transmit the signals from extracellular environment and then make further responses through changes in locations, shapes or morphologies. Common fluorescent membrane markers are not well suited for long time tracking due to their shorter retention time inside plasma membranes and/or their lower photostability. To this end, we develop a new bipolar marker, Mem-SQAC, which can stably insert into plasma membranes of different cells and exhibits a long retention time over 30 min. Mem-SQAC also inherits excellent photostability from the BODIPY dye family. Large two-photon absorption cross sections and long wavelength fluorescence emissions further enhance the competitiveness of Mem-SQAC as a membrane marker. By using Mem-SQAC, significant morphological changes of plasma membranes have been monitored during heavy metal poisoning and drug induced apoptosis of MCF-7 cells; the change tendencies are so distinctly different from each other that they can be used as indicators to distinguish different cell injuries. Further on, the complete processes of endocytosis toward Staphylococcus aureus and Escherichia coli by RAW 264.7 cells have been dynamically tracked. It is discovered that plasma membranes take quite different actions in response to the two bacteria, information unavailable in previous research reports.
Collapse
Affiliation(s)
- Xinfu Zhang
- State Key Laboratory of Fine Chemicals and ‡School of Life Science and Technology, Dalian University of Technology , Dalian 116024, China
| | | | | | | | | |
Collapse
|
87
|
Methuosis: nonapoptotic cell death associated with vacuolization of macropinosome and endosome compartments. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1630-42. [PMID: 24726643 DOI: 10.1016/j.ajpath.2014.02.028] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/30/2014] [Accepted: 02/04/2014] [Indexed: 12/19/2022]
Abstract
Apoptosis is the most widely recognized form of physiological programmed cell death. During the past three decades, various nonapoptotic forms of cell death have gained increasing attention, largely because of their potential importance in pathological processes, toxicology, and cancer therapy. A recent addition to the panoply of cell death phenotypes is methuosis. The neologism is derived from the Greek methuo (to drink to intoxication) because the hallmark of this form of cell death is displacement of the cytoplasm by large fluid-filled vacuoles derived from macropinosomes. The demise of the cell resembles many forms of necrosis, insofar as there is a loss of metabolic capacity and plasma membrane integrity, without the cell shrinkage and nuclear fragmentation associated with apoptosis. Methuosis was initially defined in glioblastoma cells after ectopic expression of activated Ras, but recent reports have described small molecules that can induce the features of methuosis in a broad spectrum of cancer cells, including those that are resistant to conventional apoptosis-inducing drugs. This review summarizes the available information about the distinguishing morphological characteristics and underlying mechanisms of methuosis. We compare and contrast methuosis with other cytopathological conditions in which accumulation of clear cytoplasmic vacuoles is a prominent feature. Finally, we highlight key questions that need to be answered to determine whether methuosis truly represents a unique form of regulated cell death.
Collapse
|
88
|
Kitambi SS, Toledo EM, Usoskin D, Wee S, Harisankar A, Svensson R, Sigmundsson K, Kalderén C, Niklasson M, Kundu S, Aranda S, Westermark B, Uhrbom L, Andäng M, Damberg P, Nelander S, Arenas E, Artursson P, Walfridsson J, Forsberg Nilsson K, Hammarström LGJ, Ernfors P. RETRACTED: Vulnerability of glioblastoma cells to catastrophic vacuolization and death induced by a small molecule. Cell 2014; 157:313-328. [PMID: 24656405 DOI: 10.1016/j.cell.2014.02.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/18/2013] [Accepted: 02/06/2014] [Indexed: 12/25/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.
Collapse
Affiliation(s)
- Satish Srinivas Kitambi
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Enrique M Toledo
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dmitry Usoskin
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Shimei Wee
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Aditya Harisankar
- Department of Medicine, HERM, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Richard Svensson
- Department of Pharmacy, UDOPP, Chemical Biology Consortium Sweden, Uppsala University, 751 05 Uppsala, Sweden
| | - Kristmundur Sigmundsson
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Christina Kalderén
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mia Niklasson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Soumi Kundu
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Sergi Aranda
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Bengt Westermark
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lene Uhrbom
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Andäng
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Damberg
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Sven Nelander
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Per Artursson
- Department of Pharmacy, UDOPP, Chemical Biology Consortium Sweden, Uppsala University, 751 05 Uppsala, Sweden
| | - Julian Walfridsson
- Department of Medicine, HERM, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Karin Forsberg Nilsson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lars G J Hammarström
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
89
|
Inflammatory stimuli reprogram macrophage phagocytosis to macropinocytosis for the rapid elimination of pathogens. PLoS Pathog 2014; 10:e1003879. [PMID: 24497827 PMCID: PMC3907376 DOI: 10.1371/journal.ppat.1003879] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 11/18/2013] [Indexed: 12/17/2022] Open
Abstract
Following an infectious challenge, macrophages have to be activated in order to allow efficient clearance of infectious pathogens, but how macrophage activation is coupled to increased clearance remains largely unknown. We here describe that inflammatory stimuli induced the reprogramming of the macrophage endocytic machinery from receptor-mediated phagocytosis to macropinocytosis, allowing the rapid transfer of internalized cargo to lysosomes in a receptor-independent manner. Reprogramming occurred through protein kinase C-mediated phosphorylation of the macrophage protein coronin 1, thereby activating phosphoinositol (PI)-3-kinase activity necessary for macropinocytic uptake. Expression of a phosphomimetic form of coronin 1 was sufficient to induce PI3-kinase activation and macropinocytosis even in the absence of inflammatory stimuli. Together these results suggest a hitherto unknown mechanism to regulate the internalization and degradation of infectious material during inflammation. The main cells that are involved in cleaning up microbial pathogens are macrophages. Upon an infection, macrophages are being recruited to the site of infection by a number of different stimuli. In addition, during an infection, macrophages are also activated by cytokines such as interferon-γ and tumor necrosis factor-α that is released from other immune cells. Such macrophage activation is important to achieve a rapid and efficient clearance of microbial pathogens. In this study, we found that macrophage activation induces uptake through macropinocytosis rather than receptor-mediate phagocytosis. As a consequence, microbial material as well as particles can be internalized far more efficiently; In addition, the internalized cargo is rapidly destroyed within lysosomes. We also provide the mechanisms for the switch from phagocytosis to macropinocytosis, which turned out to be the cytokine-induced phosphorylation of the host protein coronin 1. Phosphorylated coronin 1 activated the lipid kinase phosphoinositide 3-kinase, which is known to be responsible for the entry of cargo through macropinocytosis. Together these results provide evidence for a hitherto unrecognized mechanisms to regulate the internalization and degradation of infectious material during an infection.
Collapse
|
90
|
Sandvig K, Bergan J, Kavaliauskiene S, Skotland T. Lipid requirements for entry of protein toxins into cells. Prog Lipid Res 2014; 54:1-13. [PMID: 24462587 DOI: 10.1016/j.plipres.2014.01.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 01/05/2023]
Abstract
The plant toxin ricin and the bacterial toxin Shiga toxin both belong to a group of protein toxins having one moiety that binds to the cell surface, and another, enzymatically active moiety, that enters the cytosol and inhibits protein synthesis by inactivating ribosomes. Both toxins travel all the way from the cell surface to endosomes, the Golgi apparatus and the ER before the ribosome-inactivating moiety enters the cytosol. Shiga toxin binds to the neutral glycosphingolipid Gb3 at the cell surface and is therefore dependent on this lipid for transport into the cells, whereas ricin binds both glycoproteins and glycolipids with terminal galactose. The different steps of transport used by these toxins have specific requirements for lipid species, and with the recent developments in mass spectrometry analysis of lipids and microscopical and biochemical dissection of transport in cells, we are starting to see the complexity of endocytosis and intracellular transport. In this article we describe lipid requirements and the consequences of lipid changes for the entry and intoxication with ricin and Shiga toxin. These toxins can be a threat to human health, but can also be exploited for diagnosis and therapy, and have proven valuable as tools to study intracellular transport.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Jonas Bergan
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | - Simona Kavaliauskiene
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Tore Skotland
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
91
|
Hinton TM, Grusche F, Acharya D, Shukla R, Bansal V, Waddington LJ, Monaghan P, Muir BW. Bicontinuous cubic phase nanoparticle lipid chemistry affects toxicity in cultured cells. Toxicol Res (Camb) 2014. [DOI: 10.1039/c3tx50075f] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
92
|
Sundaramoorthy V, Walker AK, Yerbury J, Soo KY, Farg MA, Hoang V, Zeineddine R, Spencer D, Atkin JD. Extracellular wildtype and mutant SOD1 induces ER-Golgi pathology characteristic of amyotrophic lateral sclerosis in neuronal cells. Cell Mol Life Sci 2013; 70:4181-95. [PMID: 23765103 PMCID: PMC11113712 DOI: 10.1007/s00018-013-1385-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/03/2013] [Accepted: 05/23/2013] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing neurodegenerative disorder and the majority of ALS is sporadic, where misfolding and aggregation of Cu/Zn-superoxide dismutase (SOD1) is a feature shared with familial mutant-SOD1 cases. ALS is characterized by progressive neurospatial spread of pathology among motor neurons, and recently the transfer of extracellular, aggregated mutant SOD1 between cells was demonstrated in culture. However, there is currently no evidence that uptake of SOD1 into cells initiates neurodegenerative pathways reminiscent of ALS pathology. Similarly, whilst dysfunction to the ER-Golgi compartments is increasingly implicated in the pathogenesis of both sporadic and familial ALS, it remains unclear whether misfolded, wildtype SOD1 triggers ER-Golgi dysfunction. In this study we show that both extracellular, native wildtype and mutant SOD1 are taken up by macropinocytosis into neuronal cells. Hence uptake does not depend on SOD1 mutation or misfolding. We also demonstrate that purified mutant SOD1 added exogenously to neuronal cells inhibits protein transport between the ER-Golgi apparatus, leading to Golgi fragmentation, induction of ER stress and apoptotic cell death. Furthermore, we show that extracellular, aggregated, wildtype SOD1 also induces ER-Golgi pathology similar to mutant SOD1, leading to apoptotic cell death. Hence extracellular misfolded wildtype or mutant SOD1 induce dysfunction to ER-Golgi compartments characteristic of ALS in neuronal cells, implicating extracellular SOD1 in the spread of pathology among motor neurons in both sporadic and familial ALS.
Collapse
Affiliation(s)
- Vinod Sundaramoorthy
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
| | - Adam K. Walker
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
- Center for Neurodegenerative Disease Research, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Justin Yerbury
- School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Kai Ying Soo
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
| | - Manal A. Farg
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
| | - Vy Hoang
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
| | - Rafaa Zeineddine
- School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Damian Spencer
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
| | - Julie D. Atkin
- Department of Biochemistry, Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086 Australia
- Department of Florey Neuroscience, University of Melbourne, Parkville, VIC 3010 Australia
| |
Collapse
|
93
|
Amir D, Fessler DMT. Boots for Achilles: progesterone's reduction of cholesterol is a second-order adaptation. QUARTERLY REVIEW OF BIOLOGY 2013; 88:97-116. [PMID: 23909226 DOI: 10.1086/670528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Progesterone and cholesterol are both vital to pregnancy. Among other functions, progesterone downregulates inflammatory responses, allowing for maternal immune tolerance of the fetal allograft. Cholesterol a key component of cell membranes, is important in intracellular transport, cell signaling, nerve conduction, and metabolism Despite the importance of each substance in pregnancy, one exercises an antagonistic effect on the other, as periods of peak progesterone correspond with reductions in cholesterol availability, a consequence of progesterone's negative effects on cholesterol biosynthesis. This arrangement is understandable in light of the threat posed by pathogens early in pregnancy. Progesterone-induced immunomodulation entails increased vulnerability to infection, an acute problem in the first trimester, when fetal development is highly susceptible to insult. Many pathogens rely on cholesterol for cell entry, egress, and replication. Progesterone's antagonistic effects on cholesterol thus partially compensate for the costs entailed by progesterone-induced immunomodulation. Among pathogens to which the host's vulnerability is increased by progesterone's effects, approximately 90% utilize cholesterol, and this is notably true of pathogens that pose a risk during pregnancy. In addition to having a number of possible clinical applications, our approach highlights the potential importance of second-order adaptations, themselves a consequence of the lack of teleology in evolutionary processes.
Collapse
Affiliation(s)
- Dorsa Amir
- Center for Behavior, Evolution, and Culture, Department of Anthropology, University of California, Los Angeles Los Angeles, California 90095-1553, USA.
| | | |
Collapse
|
94
|
Basquin C, Sauvonnet N. Phosphoinositide 3-kinase at the crossroad between endocytosis and signaling of cytokine receptors. Commun Integr Biol 2013; 6:e24243. [PMID: 23986799 PMCID: PMC3737753 DOI: 10.4161/cib.24243] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 12/24/2022] Open
Abstract
Class I phosphoinositide 3-kinase (PI3K) is a lipid kinase playing key roles in many signaling pathways regulating cell survival and growth. Besides its important role in signal transduction, PI3K is also involved in actin and membrane reorganization such as protrusion, adhesion, phagocytosis and macropinocytosis. Receptor-mediated endocytosis is initiated by plasma membrane reorganization creating buds that then mature to small vesicles. Whereas most of endocytic mechanisms involve actin polymerization, PI3K requirement has not been clearly investigated. Our study identifies class I PI3K as a key player in clathrin-independent endocytosis of the interleukin 2 receptor (IL-2R) in contrast to the clathrin-dependent entry of transferrin (Tf). IL-2R is a cytokine receptor, inducing several signaling cascades such as PI3K, that are essential for the immune response. We have shown previously that IL-2R can be internalized with or without IL-2 and this process requires dynamin, actin and their regulators cortactin, N-WASP, Rac1 and the kinases Pak. Our recent work reveals that PI3K regulates Rac1 during IL-2R uptake in two ways: via its catalytic activity (p110) and via its regulatory factor (p85). Indeed, the catalytic activity of PI3K is required for both constitutive and IL-2 induced uptake of cytokine receptors, in lymphocytes as well as in epithelial cells. Interestingly, Vav2, a Rac1 GTPase exchange factor (GEF) induced upon PI3K activation, is specifically involved and recruited during IL-2R uptake. The second action of PI3K is via its regulatory subunit, p85, which binds activated Rac1 and IL-2R; this interaction being enhanced upon IL-2 treatment. Thus, PI3K regulates both the activation of Rac1 and its recruitment during IL-2R endocytosis. Finally, our results identify a link between cytokine receptors signaling and clathrin-independent endocytosis.
Collapse
Affiliation(s)
- Cyril Basquin
- Institut Pasteur; Unité de Biologie des Interactions Cellulaires; Paris, France ; CNRS URA 2582; Dynamique des Interactions Cellulaires; Paris, France
| | | |
Collapse
|
95
|
Chapuy-Regaud S, Subra C, Requena M, de Medina P, Amara S, Delton-Vandenbroucke I, Payre B, Cazabat M, Carriere F, Izopet J, Poirot M, Record M. Progesterone and a phospholipase inhibitor increase the endosomal bis(monoacylglycero)phosphate content and block HIV viral particle intercellular transmission. Biochimie 2013; 95:1677-88. [PMID: 23774297 DOI: 10.1016/j.biochi.2013.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/27/2013] [Indexed: 10/26/2022]
Abstract
Progesterone, the cationic amphiphile U18666A and a phospholipase inhibitor (Methyl Arachidonyl Fluoro Phosphonate, MAFP) inhibited by 70%-90% HIV production in viral reservoir cells, i.e. human THP-1 monocytes and monocyte-derived macrophages (MDM). These compounds triggered an inhibition of fluid phase endocytosis (macropinocytosis) and modified cellular lipid homeostasis since endosomes accumulated filipin-stained sterols and Bis(Monoacylglycero)Phosphate (BMP). BMP was quantified using a new cytometry procedure and was increased by 1.25 times with MAFP, 1.7 times with U18666A and 2.5 times with progesterone. MAFP but not progesterone or U18666A inhibited the hydrolysis of BMP by the Pancreatic Lipase Related Protein 2 (PLRP2) as shown by in-vitro experiments. The possible role of sterol transporters in steroid-mediated BMP increase is discussed. Electron microscopy showed the accumulation of viral particles either into large intracellular viral-containing compartments or outside the cells, indicating that endosomal accumulation of BMP could block intracellular biogenesis of viral particles while inhibition of macropinocytosis would prevent viral particle uptake. This is the first report linking BMP metabolism with a natural steroid such as progesterone or with involvement of a phospholipase A1 activity. BMP cellular content could be used as a biomarker for efficient anti-viral drugs.
Collapse
Affiliation(s)
- Sabine Chapuy-Regaud
- INSERM, U1043, Equipe Infection virales: persistance, réponse de l'hôte et physiopathologie, Toulouse F-31300, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Zubris KAV, Liu R, Colby A, Schulz MD, Colson YL, Grinstaff MW. In vitro activity of Paclitaxel-loaded polymeric expansile nanoparticles in breast cancer cells. Biomacromolecules 2013; 14:2074-82. [PMID: 23617223 PMCID: PMC3915286 DOI: 10.1021/bm400434h] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Through a series of in vitro studies, the essential steps for intracellular drug delivery of paclitaxel using a pH-responsive nanoparticle system have been investigated in breast cancer cells. We successfully encapsulated paclitaxel within polymeric expansile nanoparticles (Pax-eNPs) at 5% loading via a miniemulsion polymerization procedure. Fluorescently tagged eNPs were readily taken up by MDA-MB-231 breast cancer cells grown in culture as confirmed by confocal microscopy and flow cytometry. The ability of the encapsulated paclitaxel to reach the cytoplasm was also observed using confocal microscopy and fluorescently labeled paclitaxel. Pax-eNPs were shown to be efficacious against three in vitro human breast adenocarcinoma cell lines (MDA-MB-231, MCF-7, and SK-BR-3) as well as cells isolated from the pleural effusions of two different breast cancer patients. Lastly, macropinocytosis was identified as the major cellular pathway responsible for eNP uptake, as confirmed using temperature-sensitive metabolic reduction, pharmacologic inhibitors, and fluid-phase marker colocalization.
Collapse
Affiliation(s)
- Kimberly Ann V. Zubris
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA 02215
| | - Rong Liu
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115
| | - Aaron Colby
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA 02215
| | - Morgan D. Schulz
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA 02215
| |
Collapse
|
97
|
Endocytosis of gene delivery vectors: from clathrin-dependent to lipid raft-mediated endocytosis. Mol Ther 2013; 21:1118-30. [PMID: 23587924 DOI: 10.1038/mt.2013.54] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The ideal nonviral vector delivers its nucleic acid cargo to a specific intracellular target. Vectors enter cells mainly through endocytosis and are distributed to various intracellular organelles. Recent advances in microscopy, lipidomics, and proteomics confirm that the cell membrane is composed of clusters of lipids, organized in the form of lipid raft domains, together with non-raft domains that comprise a generally disordered lipid milieu. The binding of a nonviral vector to either region can determine the pathway for its endocytic uptake and subsequent intracellular itinerary. Given this model of the cell membrane structure, endocytic pathways should be reclassified in relation to lipid rafts. In this review, we attempt to assess the currently recognized endocytic pathways in mammalian cells. The endocytic pathways are classified in relation to the membrane regions that make up the primary endocytic vesicles. This review covers the well-recognized clathrin-mediated endocytosis (CME), phagocytosis, and macropinocytosis in addition to the less addressed pathways that take place in lipid rafts. These include caveolae-mediated, flotillin-dependent, GTPase regulator associated with focal adhesion kinase-1 (GRAF1)-dependent, adenosine diphosphate-ribosylation factor 6 (Arf6)-dependent, and RhoA-dependent endocytic pathways. We summarize the regulators associated with each uptake pathway and methods for interfering with these regulators are discussed. The fate of endocytic vesicles resulting from each endocytic uptake pathway is highlighted.
Collapse
|
98
|
Bohdanowicz M, Grinstein S. Role of Phospholipids in Endocytosis, Phagocytosis, and Macropinocytosis. Physiol Rev 2013; 93:69-106. [DOI: 10.1152/physrev.00002.2012] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Endocytosis, phagocytosis, and macropinocytosis are fundamental processes that enable cells to sample their environment, eliminate pathogens and apoptotic bodies, and regulate the expression of surface components. While a great deal of effort has been devoted over many years to understanding the proteins involved in these processes, the important contribution of phospholipids has only recently been appreciated. This review is an attempt to collate and analyze the rapidly emerging evidence documenting the role of phospholipids in clathrin-mediated endocytosis, phagocytosis, and macropinocytosis. A primer on phospholipid biosynthesis, catabolism, subcellular distribution, and transport is presented initially, for reference, together with general considerations of the effects of phospholipids on membrane curvature and charge. This is followed by a detailed analysis of the critical functions of phospholipids in the internalization processes and in the maturation of the resulting vesicles and vacuoles as they progress along the endo-lysosomal pathway.
Collapse
Affiliation(s)
- Michal Bohdanowicz
- Division of Cell Biology, Hospital for Sick Children, and Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Sergio Grinstein
- Division of Cell Biology, Hospital for Sick Children, and Institute of Medical Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
99
|
Faille D, El-Assaad F, Mitchell AJ, Alessi MC, Chimini G, Fusai T, Grau GE, Combes V. Endocytosis and intracellular processing of platelet microparticles by brain endothelial cells. J Cell Mol Med 2012; 16:1731-8. [PMID: 21883894 PMCID: PMC3822686 DOI: 10.1111/j.1582-4934.2011.01434.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Platelet-derived microparticles (PMP) bind and modify the phenotype of many cell types including endothelial cells. Recently, we showed that PMP were internalized by human brain endothelial cells (HBEC). Here we intend to better characterize the internalization mechanisms of PMP and their intracellular fate. Confocal microscopy analysis of PKH67-labelled PMP distribution in HBEC showed PMP in early endosome antigen 1 positive endosomes and in LysoTracker-labelled lysosomes, confirming a role for endocytosis in PMP internalization. No fusion of calcein-loaded PMP with HBEC membranes was observed. Quantification of PMP endocytosis using flow cytometry revealed that it was partially inhibited by trypsin digestion of PMP surface proteins and by extracellular Ca2+ chelation by EDTA, suggesting a partial role for receptor-mediated endocytosis in PMP uptake. This endocytosis was independent of endothelial receptors such as intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 and was not increased by tumour necrosis factor stimulation of HBEC. Platelet-derived microparticle internalization was dramatically increased in the presence of decomplemented serum, suggesting a role for PMP opsonin-dependent phagocytosis. Platelet-derived microparticle uptake was greatly diminished by treatment of HBEC with cytochalasin D, an inhibitor of microfilament formation required for both phagocytosis and macropinocytosis, with methyl-β-cyclodextrin that depletes membrane cholesterol needed for macropinocytosis and with amiloride that inhibits the Na+/H+ exchanger involved in macropinocytosis. In conclusion, PMP are taken up by active endocytosis in HBEC, involving mechanisms consistent with both phagocytosis and macropinocytosis. These findings identify new processes by which PMP could modify endothelial cell phenotype and functions.
Collapse
Affiliation(s)
- Dorothée Faille
- Department of Pathology, University of Sydney, Camperdown, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Butler CE, Tyler KM. Membrane traffic and synaptic cross-talk during host cell entry by Trypanosoma cruzi. Cell Microbiol 2012; 14:1345-53. [PMID: 22646288 PMCID: PMC3428839 DOI: 10.1111/j.1462-5822.2012.01818.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 05/23/2012] [Accepted: 05/23/2012] [Indexed: 12/24/2022]
Abstract
It is widely accepted that Trypanosoma cruzi can exploit the natural exocytic response of the host to cell damage, utilizing host cell lysosomes as important effectors. It is, though, increasingly clear that the parasite also exploits endocytic mechanisms which allow for incorporation of plasma membrane into the parasitophorous vacuole. Further, that these endocytic mechanisms are involved in cross-talk with the exocytic machinery, in the recycling of vesicles and in the manipulation of the cytoskeleton. Here we review the mechanisms by which T. cruzi exploits features of the exocytic and endocytic pathways in epithelial and endothelial cells and the evidence for cross-talk between these pathways.
Collapse
Affiliation(s)
- Claire E Butler
- Biomedical Research Centre, Norwich School of Medicine, University of East Anglia, Norwich, NR4 7TJ, UK
| | | |
Collapse
|