51
|
Rueda-Gensini L, Cifuentes J, Castellanos MC, Puentes PR, Serna JA, Muñoz-Camargo C, Cruz JC. Tailoring Iron Oxide Nanoparticles for Efficient Cellular Internalization and Endosomal Escape. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1816. [PMID: 32932957 PMCID: PMC7559083 DOI: 10.3390/nano10091816] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Iron oxide nanoparticles (IONs) have been widely explored for biomedical applications due to their high biocompatibility, surface-coating versatility, and superparamagnetic properties. Upon exposure to an external magnetic field, IONs can be precisely directed to a region of interest and serve as exceptional delivery vehicles and cellular markers. However, the design of nanocarriers that achieve an efficient endocytic uptake, escape lysosomal degradation, and perform precise intracellular functions is still a challenge for their application in translational medicine. This review highlights several aspects that mediate the activation of the endosomal pathways, as well as the different properties that govern endosomal escape and nuclear transfection of magnetic IONs. In particular, we review a variety of ION surface modification alternatives that have emerged for facilitating their endocytic uptake and their timely escape from endosomes, with special emphasis on how these can be manipulated for the rational design of cell-penetrating vehicles. Moreover, additional modifications for enhancing nuclear transfection are also included in the design of therapeutic vehicles that must overcome this barrier. Understanding these mechanisms opens new perspectives in the strategic development of vehicles for cell tracking, cell imaging and the targeted intracellular delivery of drugs and gene therapy sequences and vectors.
Collapse
Affiliation(s)
- Laura Rueda-Gensini
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Javier Cifuentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Maria Claudia Castellanos
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Julian A. Serna
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
52
|
The Role of Receptor Tyrosine Kinases in Lassa Virus Cell Entry. Viruses 2020; 12:v12080857. [PMID: 32781509 PMCID: PMC7472032 DOI: 10.3390/v12080857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 02/06/2023] Open
Abstract
The zoonotic Old World mammarenavirus Lassa (LASV) causes severe hemorrhagic fever with high mortality and morbidity in humans in endemic regions. The development of effective strategies to combat LASV infections is of high priority, given the lack of a licensed vaccine and restriction on available treatment to off-label use of ribavirin. A better understanding of the fundamental aspects of the virus's life cycle would help to improve the development of novel therapeutic approaches. Host cell entry and restriction factors represent major barriers for emerging viruses and are promising targets for therapeutic intervention. In addition to the LASV main receptor, the extracellular matrix molecule dystroglycan (DG), the phosphatidylserine-binding receptors of the Tyro3/Axl/Mer (TAM), and T cell immunoglobulin and mucin receptor (TIM) families are potential alternative receptors of LASV infection. Therefore, the relative contributions of candidate receptors to LASV entry into a particular human cell type are a complex function of receptor expression and functional DG availability. Here, we describe the role of two receptor tyrosine kinases (RTKs), Axl and hepatocyte growth factor receptor (HGFR), in the presence and absence of glycosylated DG for LASV entry. We found that both RTKs participated in the macropinocytosis-related LASV entry and, regardless of the presence or absence of functional DG, their inhibition resulted in a significant antiviral effect.
Collapse
|
53
|
Polysialic acid and Siglec-E orchestrate negative feedback regulation of microglia activation. Cell Mol Life Sci 2020; 78:1637-1653. [PMID: 32725371 PMCID: PMC7904730 DOI: 10.1007/s00018-020-03601-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 11/09/2022]
Abstract
Polysialic acid (polySia) emerges as a novel regulator of microglia activity. We recently identified polysialylated proteins in the Golgi compartment of murine microglia that are released in response to inflammatory stimulation. Since exogenously added polySia is able to attenuate the inflammatory response, we proposed that the release of polysialylated proteins constitutes a mechanism for negative feedback regulation of microglia activation. Here, we demonstrate that translocation of polySia from the Golgi to the cell surface can be induced by calcium depletion of the Golgi compartment and that polysialylated proteins are continuously released for at least 24 h after the onset of inflammatory stimulation. The latter was unexpected, because polySia signals detected by immunocytochemistry are rapidly depleted. However, it indicates that the amount of released polySia is much higher than anticipated based on immunostaining. This may be crucial for microglial responses during traumatic brain injury (TBI), as we detected polySia signals in activated microglia around a stab wound in the adult mouse brain. In BV2 microglia, the putative polySia receptor Siglec-E is internalized during lipopolysaccharide (LPS)-induced activation and in response to polySia exposure, indicating interaction. Correspondingly, CRISPR/Cas9-mediated Siglec-E knockout prevents inhibition of pro inflammatory activation by exogenously added polySia and leads to a strong increase of the LPS response. A comparable increase of LPS-induced activation has been observed in microglia with abolished polySia synthesis. Together, these results indicate that the release of the microglia-intrinsic polySia pool, as implicated in TBI, inhibits the inflammatory response by acting as a trans-activating ligand of Siglec-E.
Collapse
|
54
|
Abstract
My coworkers and I have used animal viruses and their interaction with host cells to investigate cellular processes difficult to study by other means. This approach has allowed us to branch out in many directions, including membrane protein characterization, endocytosis, secretion, protein folding, quality control, and glycobiology. At the same time, our aim has been to employ cell biological approaches to expand the fundamental understanding of animal viruses and their pathogenic lifestyles. We have studied mechanisms of host cell entry and the uncoating of incoming viruses as well as the synthesis, folding, maturation, and intracellular movement of viral proteins and molecular assemblies. I have had the privilege to work in institutions in four different countries. The early years in Finland (the University of Helsinki) were followed by 6 years in Germany (European Molecular Biology Laboratory), 16 years in the United States (Yale School of Medicine), and 16 years in Switzerland (ETH Zurich).
Collapse
Affiliation(s)
- Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland;
| |
Collapse
|
55
|
Abstract
Several studies have demonstrated interactions between the two leaflets in membrane bilayers and the importance of specific lipid species for such interaction and membrane function. We here discuss these investigations with a focus on the sphingolipid and cholesterol-rich lipid membrane domains called lipid rafts, including the small flask-shaped invaginations called caveolae, and the importance of such membrane structures in cell biology and cancer. We discuss the possible interactions between the very long-chain sphingolipids in the outer leaflet of the plasma membrane and the phosphatidylserine species PS 18:0/18:1 in the inner leaflet and the importance of cholesterol for such interactions. We challenge the view that lipid rafts contain a large fraction of lipids with two saturated fatty acyl groups and argue that it is important in future studies of membrane models to use asymmetric membrane bilayers with lipid species commonly found in cellular membranes. We also discuss the need for more quantitative lipidomic studies in order to understand membrane function and structure in general, and the importance of lipid rafts in biological systems. Finally, we discuss cancer-related changes in lipid rafts and lipid composition, with a special focus on changes in glycosphingolipids and the possibility of using lipid therapy for cancer treatment.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway.
| |
Collapse
|
56
|
Zhang L, Wang F, Li Q, Wang L, Fan C, Li J, Zhu Y. Classifying Cell Types with DNA-Encoded Ligand-Receptor Interactions on the Cell Membrane. NANO LETTERS 2020; 20:3521-3527. [PMID: 32223268 DOI: 10.1021/acs.nanolett.0c00445] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clustering, endocytosis, and intracellular transport of molecules on the cell membrane are critically dependent on the type of cells. However, the membrane-associated redistribution of molecules has not been exploited to realize cell classification for diagnostic purposes. Here, we develop a set of DNA-encoded artificial receptors and ligands to monitor the cell membrane redistribution. In this system, a cholesterol-modified single-stranded DNA strand serves as the receptor localized on the membrane, and a tetrahedral DNA framework (TDF) nanostructure with a complementary overhang serves as the ligand. The DNA-encoded receptor-ligand interaction is highly orthogonal, mimicking the dynamics of natural receptors and ligands on cells. We demonstrate that the dynamics of membrane redistribution can be resolved by the dual-color fluorescent patterns of the receptor-ligand interactions in a single image, which can be exploited to classify cell lines with high fidelity. This DNA-encoded method thus holds great promise for cell typing and diagnosis.
Collapse
Affiliation(s)
- Luhao Zhang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Qian Li
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Chunhai Fan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Jiang Li
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| |
Collapse
|
57
|
Multifaceted Functions of Host Cell Caveolae/Caveolin-1 in Virus Infections. Viruses 2020; 12:v12050487. [PMID: 32357558 PMCID: PMC7291293 DOI: 10.3390/v12050487] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
Virus infection has drawn extensive attention since it causes serious or even deadly diseases, consequently inducing a series of social and public health problems. Caveolin-1 is the most important structural protein of caveolae, a membrane invagination widely known for its role in endocytosis and subsequent cytoplasmic transportation. Caveolae/caveolin-1 is tightly associated with a wide range of biological processes, including cholesterol homeostasis, cell mechano-sensing, tumorigenesis, and signal transduction. Intriguingly, the versatile roles of caveolae/caveolin-1 in virus infections have increasingly been appreciated. Over the past few decades, more and more viruses have been identified to invade host cells via caveolae-mediated endocytosis, although other known pathways have been explored. The subsequent post-entry events, including trafficking, replication, assembly, and egress of a large number of viruses, are caveolae/caveolin-1-dependent. Deprivation of caveolae/caveolin-1 by drug application or gene editing leads to abnormalities in viral uptake, viral protein expression, or virion release, whereas the underlying mechanisms remain elusive and must be explored holistically to provide potential novel antiviral targets and strategies. This review recapitulates our current knowledge on how caveolae/caveolin-1 functions in every step of the viral infection cycle and various relevant signaling pathways, hoping to provide a new perspective for future viral cell biology research.
Collapse
|
58
|
Cold Atmospheric Plasma Stimulates Clathrin-Dependent Endocytosis to Repair Oxidised Membrane and Enhance Uptake of Nanomaterial in Glioblastoma Multiforme Cells. Sci Rep 2020; 10:6985. [PMID: 32332819 PMCID: PMC7181794 DOI: 10.1038/s41598-020-63732-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/25/2020] [Indexed: 01/16/2023] Open
Abstract
Cold atmospheric plasma (CAP) enhances uptake and accumulation of nanoparticles and promotes synergistic cytotoxicity against cancer cells. However, the mechanisms are not well understood. In this study, we investigate the enhanced uptake of theranostic nanomaterials by CAP. Numerical modelling of the uptake of gold nanoparticle into U373MG Glioblastoma multiforme (GBM) cells predicts that CAP may introduce a new uptake route. We demonstrate that cell membrane repair pathways play the main role in this stimulated new uptake route, following non-toxic doses of dielectric barrier discharge CAP. CAP treatment induces cellular membrane damage, mainly via lipid peroxidation as a result of reactive oxygen species (ROS) generation. Membranes rich in peroxidised lipids are then trafficked into cells via membrane repairing endocytosis. We confirm that the enhanced uptake of nanomaterials is clathrin-dependent using chemical inhibitors and silencing of gene expression. Therefore, CAP-stimulated membrane repair increases endocytosis and accelerates the uptake of gold nanoparticles into U373MG cells after CAP treatment. We demonstrate the utility of CAP to model membrane oxidative damage in cells and characterise a previously unreported mechanism of membrane repair to trigger nanomaterial uptake. This knowledge will underpin the development of new delivery strategies for theranostic nanoparticles into cancer cells.
Collapse
|
59
|
Abstract
Transcytosis of macromolecules through lung endothelial cells is the primary route of transport from the vascular compartment into the interstitial space. Endothelial transcytosis is mostly a caveolae-dependent process that combines receptor-mediated endocytosis, vesicle trafficking via actin-cytoskeletal remodeling, and SNARE protein directed vesicle fusion and exocytosis. Herein, we review the current literature on caveolae-mediated endocytosis, the role of actin cytoskeleton in caveolae stabilization at the plasma membrane, actin remodeling during vesicle trafficking, and exocytosis of caveolar vesicles. Next, we provide a concise summary of experimental methods employed to assess transcytosis. Finally, we review evidence that transcytosis contributes to the pathogenesis of acute lung injury. © 2020 American Physiological Society. Compr Physiol 10:491-508, 2020.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Correspondence to
| |
Collapse
|
60
|
Yuan X, Qin B, Yin H, Shi Y, Jiang M, Luo L, Luo Z, Zhang J, Li X, Zhu C, Du Y, You J. Virus-like Nonvirus Cationic Liposome for Efficient Gene Delivery via Endoplasmic Reticulum Pathway. ACS CENTRAL SCIENCE 2020; 6:174-188. [PMID: 32123735 PMCID: PMC7047280 DOI: 10.1021/acscentsci.9b01052] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Indexed: 05/11/2023]
Abstract
Gene vectors play a critical role in gene therapy. To achieve efficient transfection, we developed a novel nonvirus cationic liposome (Lipo-Par), which was bound covalently with the cationic polypeptide pardaxin (Par). Interestingly, the Lipo-Pars exhibited highly enhanced gene transfection efficiency in various cell lines compared to that of the non-Par-bonded liposomes (Lipo-Nons). As a result, the internalization and intracellular transport mechanisms of the Lipo-Pars were investigated, and the findings indicated their ability to actively target the endoplasmic reticulum (ER) by moving along the cell cytoskeleton after undergoing caveolin-mediated endocytosis. This intracellular transport process is similar to that of some viruses. It was also found that ER stress and calcium level disturbances can affect the Lipo-Par-mediated expression of certain exogenous genes. A possible, yet non-negligible explanation for the high transfection efficiency of the Lipo-Par is its virus-like intracellular behavior and the intimate relationship between the ER membrane and the nuclear envelope.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jian You
- . Phone: 086-571-88208443. Fax: 086-571-88208439
| |
Collapse
|
61
|
Wang J, Li Y, Wang S, Liu F. Dynamics of transmissible gastroenteritis virus internalization unraveled by single-virus tracking in live cells. FASEB J 2020; 34:4653-4669. [PMID: 32017270 PMCID: PMC7163995 DOI: 10.1096/fj.201902455r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Transmissible gastroenteritis virus (TGEV) is a swine enteropathogenic coronavirus that causes significant economic losses in swine industry. Current studies on TGEV internalization mainly focus on viral receptors, but the internalization mechanism is still unclear. In this study, we used single‐virus tracking to obtain the detailed insights into the dynamic events of the TGEV internalization and depict the whole sequential process. We observed that TGEVs could be internalized through clathrin‐ and caveolae‐mediated endocytosis, and the internalization of TGEVs was almost completed within ~2 minutes after TGEVs attached to the cell membrane. Furthermore, the interactions of TGEVs with actin and dynamin 2 in real time during the TGEV internalization were visualized. To our knowledge, this is the first report that single‐virus tracking technique is used to visualize the entire dynamic process of the TGEV internalization: before the TGEV internalization, with the assistance of actin, clathrin, and caveolin 1 would gather around the virus to form the vesicle containing the TGEV, and after ~60 seconds, dynamin 2 would be recruited to promote membrane fission. These results demonstrate that TGEVs enter ST cells via clathrin‐ and caveolae‐mediated endocytic, actin‐dependent, and dynamin 2‐dependent pathways.
Collapse
Affiliation(s)
- Jian Wang
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, China
| | - Yangyang Li
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, China
| | - Shouyu Wang
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, China.,Computational Optics Laboratory, School of Science, Jiangnan University, Wuxi, China
| | - Fei Liu
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
62
|
Dhanda AS, Yu C, Lulic KT, Vogl AW, Rausch V, Yang D, Nichols BJ, Kim SH, Polo S, Hansen CG, Guttman JA. Listeria monocytogenes Exploits Host Caveolin for Cell-to-Cell Spreading. mBio 2020; 11:e02857-19. [PMID: 31964732 PMCID: PMC6974566 DOI: 10.1128/mbio.02857-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Listeria monocytogenes moves from one cell to another using actin-rich membrane protrusions that propel the bacterium toward neighboring cells. Despite cholesterol being required for this transfer process, the precise host internalization mechanism remains elusive. Here, we show that caveolin endocytosis is key to this event as bacterial cell-to-cell transfer is severely impaired when cells are depleted of caveolin-1. Only a subset of additional caveolar components (cavin-2 and EHD2) are present at sites of bacterial transfer, and although clathrin and the clathrin-associated proteins Eps15 and AP2 are absent from the bacterial invaginations, efficient L. monocytogenes spreading requires the clathrin-interacting protein epsin-1. We also directly demonstrated that isolated L. monocytogenes membrane protrusions can trigger the recruitment of caveolar proteins in a neighboring cell. The engulfment of these bacterial and cytoskeletal structures through a caveolin-based mechanism demonstrates that the classical nanometer-scale theoretical size limit for this internalization pathway is exceeded by these bacterial pathogens.IMPORTANCEListeria monocytogenes moves from one cell to another as it disseminates within tissues. This bacterial transfer process depends on the host actin cytoskeleton as the bacterium forms motile actin-rich membranous protrusions that propel the bacteria into neighboring cells, thus forming corresponding membrane invaginations. Here, we examine these membrane invaginations and demonstrate that caveolin-1-based endocytosis is crucial for efficient bacterial cell-to-cell spreading. We show that only a subset of caveolin-associated proteins (cavin-2 and EHD2) are involved in this process. Despite the absence of clathrin at the invaginations, the classical clathrin-associated protein epsin-1 is also required for efficient bacterial spreading. Using isolated L. monocytogenes protrusions added onto naive host cells, we demonstrate that actin-based propulsion is dispensable for caveolin-1 endocytosis as the presence of the protrusion/invagination interaction alone triggers caveolin-1 recruitment in the recipient cells. Finally, we provide a model of how this caveolin-1-based internalization event can exceed the theoretical size limit for this endocytic pathway.
Collapse
Affiliation(s)
- Aaron S Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Connie Yu
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Katarina T Lulic
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - A Wayne Vogl
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Valentina Rausch
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Diana Yang
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Sung Hyun Kim
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Simona Polo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- Dipartimento di oncologia ed emato-oncologia, Universita' degli Studi di Milano, Milan, Italy
| | - Carsten G Hansen
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
63
|
Horníková L, Bruštíková K, Forstová J. Microtubules in Polyomavirus Infection. Viruses 2020; 12:E121. [PMID: 31963741 PMCID: PMC7019765 DOI: 10.3390/v12010121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Microtubules, part of the cytoskeleton, are indispensable for intracellular movement, cell division, and maintaining cell shape and polarity. In addition, microtubules play an important role in viral infection. In this review, we summarize the role of the microtubules' network during polyomavirus infection. Polyomaviruses usurp microtubules and their motors to travel via early and late acidic endosomes to the endoplasmic reticulum. As shown for SV40, kinesin-1 and microtubules are engaged in the release of partially disassembled virus from the endoplasmic reticulum to the cytosol, and dynein apparently assists in the further disassembly of virions prior to their translocation to the cell nucleus-the place of their replication. Polyomavirus gene products affect the regulation of microtubule dynamics. Early T antigens destabilize microtubules and cause aberrant mitosis. The role of these activities in tumorigenesis has been documented. However, its importance for productive infection remains elusive. On the other hand, in the late phase of infection, the major capsid protein, VP1, of the mouse polyomavirus, counteracts T-antigen-induced destabilization. It physically binds microtubules and stabilizes them. The interaction results in the G2/M block of the cell cycle and prolonged S phase, which is apparently required for successful completion of the viral replication cycle.
Collapse
Affiliation(s)
| | | | - Jitka Forstová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, 25250 Vestec, Czech Republic; (L.H.); (K.B.)
| |
Collapse
|
64
|
Liu SL, Wang ZG, Xie HY, Liu AA, Lamb DC, Pang DW. Single-Virus Tracking: From Imaging Methodologies to Virological Applications. Chem Rev 2020; 120:1936-1979. [PMID: 31951121 PMCID: PMC7075663 DOI: 10.1021/acs.chemrev.9b00692] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Uncovering
the mechanisms of virus infection and assembly is crucial
for preventing the spread of viruses and treating viral disease. The
technique of single-virus tracking (SVT), also known as single-virus
tracing, allows one to follow individual viruses at different parts
of their life cycle and thereby provides dynamic insights into fundamental
processes of viruses occurring in live cells. SVT is typically based
on fluorescence imaging and reveals insights into previously unreported
infection mechanisms. In this review article, we provide the readers
a broad overview of the SVT technique. We first summarize recent advances
in SVT, from the choice of fluorescent labels and labeling strategies
to imaging implementation and analytical methodologies. We then describe
representative applications in detail to elucidate how SVT serves
as a valuable tool in virological research. Finally, we present our
perspectives regarding the future possibilities and challenges of
SVT.
Collapse
Affiliation(s)
- Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine , Nankai University , Tianjin 300071 , P. R. China.,Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry , China University of Geosciences , Wuhan 430074 , P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine , Nankai University , Tianjin 300071 , P. R. China
| | - Hai-Yan Xie
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , P. R. China
| | - An-An Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine , Nankai University , Tianjin 300071 , P. R. China
| | - Don C Lamb
- Physical Chemistry, Department of Chemistry, Center for Nanoscience (CeNS), and Center for Integrated Protein Science Munich (CIPSM) and Nanosystems Initiative Munich (NIM) , Ludwig-Maximilians-Universität , München , 81377 , Germany
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine , Nankai University , Tianjin 300071 , P. R. China.,College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology , Wuhan University , Wuhan 430072 , P. R. China
| |
Collapse
|
65
|
Nelemans LC, Gurevich L. Drug Delivery with Polymeric Nanocarriers-Cellular Uptake Mechanisms. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E366. [PMID: 31941006 PMCID: PMC7013754 DOI: 10.3390/ma13020366] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
Abstract
Nanocarrier-based systems hold a promise to become "Dr. Ehrlich's Magic Bullet" capable of delivering drugs, proteins and genetic materials intact to a specific location in an organism down to subcellular level. The key question, however, how a nanocarrier is internalized by cells and how its intracellular trafficking and the fate in the cell can be controlled remains yet to be answered. In this review we survey drug delivery systems based on various polymeric nanocarriers, their uptake mechanisms, as well as the experimental techniques and common pathway inhibitors applied for internalization studies. While energy-dependent endocytosis is observed as the main uptake pathway, the integrity of a drug-loaded nanocarrier upon its internalization appears to be a seldomly addressed problem that can drastically affect the uptake kinetics and toxicity of the system in vitro and in vivo.
Collapse
Affiliation(s)
| | - Leonid Gurevich
- Department of Materials and Production, Aalborg University, 9220 Aalborg, Denmark;
| |
Collapse
|
66
|
Indira Chandran V, Månsson AS, Barbachowska M, Cerezo-Magaña M, Nodin B, Joshi B, Koppada N, Saad OM, Gluz O, Isaksson K, Borgquist S, Jirström K, Nabi IR, Jernström H, Belting M. Hypoxia Attenuates Trastuzumab Uptake and Trastuzumab-Emtansine (T-DM1) Cytotoxicity through Redistribution of Phosphorylated Caveolin-1. Mol Cancer Res 2020; 18:644-656. [PMID: 31900313 DOI: 10.1158/1541-7786.mcr-19-0856] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/28/2019] [Accepted: 12/20/2019] [Indexed: 11/16/2022]
Abstract
The antibody-drug conjugate trastuzumab-emtansine (T-DM1) offers an additional treatment option for patients with HER2-amplified tumors. However, primary and acquired resistance is a limiting factor in a significant subset of patients. Hypoxia, a hallmark of cancer, regulates the trafficking of several receptor proteins with potential implications for tumor targeting. Here, we have investigated how hypoxic conditions may regulate T-DM1 treatment efficacy in breast cancer. The therapeutic effect of T-DM1 and its metabolites was evaluated in conjunction with biochemical, flow cytometry, and high-resolution imaging studies to elucidate the functional and mechanistic aspects of hypoxic regulation. HER2 and caveolin-1 expression was investigated in a well-annotated breast cancer cohort. We find that hypoxia fosters relative resistance to T-DM1 in HER2+ cells (SKBR3 and BT474). This effect was not a result of deregulated HER2 expression or resistance to emtansine and its metabolites. Instead, we show that hypoxia-induced translocation of caveolin-1 from cytoplasmic vesicles to the plasma membrane contributes to deficient trastuzumab internalization and T-DM1 chemosensitivity. Caveolin-1 depletion mimicked the hypoxic situation, indicating that vesicular caveolin-1 is indispensable for trastuzumab uptake and T-DM1 cytotoxicity. In vitro studies suggested that HER2 and caveolin-1 are not coregulated, which was supported by IHC analysis in patient tumors. We find that phosphorylation-deficient caveolin-1 inhibits trastuzumab internalization and T-DM1 cytotoxicity, suggesting a specific role for caveolin-1 phosphorylation in HER2 trafficking. IMPLICATIONS: Together, our data for the first time identify hypoxic regulation of caveolin-1 as a resistance mechanism to T-DM1 with potential implications for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Vineesh Indira Chandran
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
| | - Ann-Sofie Månsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Magdalena Barbachowska
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Myriam Cerezo-Magaña
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Björn Nodin
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Neelima Koppada
- BioAnalytical Sciences-ADT, gRED, Genentech, South San Francisco, California
| | - Ola M Saad
- BioAnalytical Sciences-ADT, gRED, Genentech, South San Francisco, California
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany
| | - Karolin Isaksson
- Division of Surgery, Lund University, Skåne University Hospital, Lund, Central Hospital, Kristianstad, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
- Department of Oncology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Karin Jirström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Ivan Robert Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Helena Jernström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Mattias Belting
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
- Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
67
|
Yang Y, Wang B, Li B. Structural Requirement of Casein Peptides for Transcytosis through the Caco-2 Cell Monolayer: Hydrophobicity and Charge Property Affect the Transport Pathway and Efficiency. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11778-11787. [PMID: 31554398 DOI: 10.1021/acs.jafc.9b04831] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Casein is a rich source of bioactive peptides with complete amino acid composition. In this study, the casein peptides identified in our previous study with different hydrophobicities and charge properties were employed to investigate the transport efficiency via the transcytosis pathway across Caco-2 cell monolayers. Results revealed that the apparent permeability coefficient (Papp) values of transcytosis exhibited a linear correlation with a pI of positively charged peptides during bidirectional transport. A similar law was found as for the peptides with different hydrophobicities. The transcytosis route of Pep-II to Pep-VII appears to be the clathrin- and caveolin-independent transcytosis pathway as well as caveolae-mediated transcytosis pathway, showing a linear correlation with Papp values, respectively. Additionally, no direct correlation was shown between the hydrophobicity of peptides and clathrin-mediated transcytosis. Our results help to increase the bioaccessibility of peptide drugs across intestinal mucosa by developing strategies to alter the physicochemical properties without changing bioactivity.
Collapse
Affiliation(s)
- Yijie Yang
- College of Food Science and Nutritional Engineering , China Agricultural University , Beijing 100083 , China
| | - Bo Wang
- College of Food Science and Nutritional Engineering , China Agricultural University , Beijing 100083 , China
| | - Bo Li
- College of Food Science and Nutritional Engineering , China Agricultural University , Beijing 100083 , China
- Key Laboratory of Functional Dairy , Ministry of Education , Beijing 100083 , China
| |
Collapse
|
68
|
Chen YJ, Liu X, Tsai B. SV40 Hijacks Cellular Transport, Membrane Penetration, and Disassembly Machineries to Promote Infection. Viruses 2019; 11:v11100917. [PMID: 31590347 PMCID: PMC6832212 DOI: 10.3390/v11100917] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
During entry, a virus must be transported through the endomembrane system of the host cell, penetrate a cellular membrane, and undergo capsid disassembly, to reach the cytosol and often the nucleus in order to cause infection. To do so requires the virus to coordinately exploit the action of cellular membrane transport, penetration, and disassembly machineries. How this is accomplished remains enigmatic for many viruses, especially for viruses belonging to the nonenveloped virus family. In this review, we present the current model describing infectious entry of the nonenveloped polyomavirus (PyV) SV40. Insights from SV40 entry are likely to provide strategies to combat PyV-induced diseases, and to illuminate cellular trafficking, membrane transport, and disassembly mechanisms.
Collapse
Affiliation(s)
- Yu-Jie Chen
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA.
| | - Xiaofang Liu
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA.
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA.
| |
Collapse
|
69
|
Pennington MR, Saha A, Painter DF, Gavazzi C, Ismail AM, Zhou X, Chodosh J, Rajaiya J. Disparate Entry of Adenoviruses Dictates Differential Innate Immune Responses on the Ocular Surface. Microorganisms 2019; 7:E351. [PMID: 31540200 PMCID: PMC6780103 DOI: 10.3390/microorganisms7090351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 12/31/2022] Open
Abstract
Human adenovirus infection of the ocular surface is associated with severe keratoconjunctivitis and the formation of subepithelial corneal infiltrates, which may persist and impair vision for months to years following infection. Long term pathology persists well beyond the resolution of viral replication, indicating that the prolonged immune response is not virus-mediated. However, it is not clear how these responses are sustained or even initiated following infection. This review discusses recent work from our laboratory and others which demonstrates different entry pathways specific to both adenovirus and cell type. These findings suggest that adenoviruses may stimulate specific pattern recognition receptors in an entry/trafficking-dependent manner, leading to distinct immune responses dependent on the virus/cell type combination. Additional work is needed to understand the specific connections between adenoviral entry and the stimulation of innate immune responses by the various cell types present on the ocular surface.
Collapse
Affiliation(s)
- Matthew R Pennington
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Amrita Saha
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - David F Painter
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Christina Gavazzi
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Ashrafali M Ismail
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Xiaohong Zhou
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - James Chodosh
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Jaya Rajaiya
- Howe Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
70
|
Hilgemann DW, Lin MJ, Fine M, Deisl C. On the existence of endocytosis driven by membrane phase separations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183007. [PMID: 31202864 DOI: 10.1016/j.bbamem.2019.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/31/2019] [Accepted: 06/06/2019] [Indexed: 01/15/2023]
Abstract
Large endocytic responses can occur rapidly in diverse cell types without dynamins, clathrin, or actin remodeling. Our experiments suggest that membrane phase separations are crucial with more ordered plasma membrane domains being internalized. Not only do these endocytic processes rely on coalescence of membrane domains, they are promoted by participation of membrane proteins in such domains, one important regulatory influence being palmitoylation. Membrane actin cytoskeleton in general resists membrane phase transitions, and its remodeling may play many roles. Besides membrane 'caging' and 'pinching' roles, typically ascribed to clathrin and dynamins, cytoskeleton remodeling may modify local membrane tension and buckling, as well as the presence and location of actin- and tension-free membrane patches. Endocytosis that depends on membrane phase separations becomes activated in metabolic stress and in response to Ca and PI3 kinase signaling. Internalized membrane traffics normally, and the secretory pathway eventually resupplies membrane to the plasmalemma or directs internalized membrane to other locations, including the extracellular space as exosomes. We describe here that endocytosis driven by membrane phase transitions is regulated by the same signaling mechanisms that regulate macropinocytosis, and it may play diverse roles in cells from nutrient assimilation to membrane recycling, cell migration, and the initiation of quiescent or hibernating cell states. Membrane ordering and phase separations have been shown to promote endocytosis in diverse cell types, including fibroblasts, myocytes, glial cells, and immune cells. We propose that clathrin/dynamin-independent endocytosis represents a continuum of related mechanisms with variable but universal dependence on membrane ordering and actin remodeling. This article is part of a Special Issue entitled: Molecular biophysics of membranes and membrane proteins.
Collapse
Affiliation(s)
- Donald W Hilgemann
- University of Texas Southwestern Medical Center, Department of Physiology, 5323 Harry Hines Boulevard, Dallas, TX 75235-9040, USA.
| | - Mei-Jung Lin
- University of Texas Southwestern Medical Center, Department of Physiology, 5323 Harry Hines Boulevard, Dallas, TX 75235-9040, USA
| | - Michael Fine
- University of Texas Southwestern Medical Center, Department of Physiology, 5323 Harry Hines Boulevard, Dallas, TX 75235-9040, USA
| | - Christine Deisl
- University of Texas Southwestern Medical Center, Department of Physiology, 5323 Harry Hines Boulevard, Dallas, TX 75235-9040, USA
| |
Collapse
|
71
|
Aliyu IA, Ling KH, Md Hashim NF, Lam JY, Chee HY. Annexin II as a Dengue Virus Serotype 2 Interacting Protein Mediating Virus Interaction on Vero Cells. Viruses 2019; 11:v11040335. [PMID: 30970587 PMCID: PMC6520844 DOI: 10.3390/v11040335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023] Open
Abstract
Recent evidence has demonstrated that dengue virus requires active filopodia formation for a successful infection. However, the cellular factor involved in the interaction has not been fully elucidated. We used a combination of virus overlay protein binding assay and LC-MS/MS, and identified annexin II as a dengue virus serotype 2 (DENV2) interacting protein on Vero cells, upon filopodia induction. Flow cytometry analysis showed annexin II on the Vero cells surface increased when DENV2 was added. The amount of annexin II in the plasma membrane fraction was reduced as the infection progressed. Antibody-mediated inhibition of infection and siRNA-mediated knockdown of annexin II expression significantly reduced DENV2 infection and production levels. Collectively, we demonstrated that annexin II is one of the host factor involved in DENV2 binding on Vero cells.
Collapse
Affiliation(s)
- Isah Abubakar Aliyu
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Department of Medical Laboratory Science, Faculty of Allied Health Science, College of Health Science, Bayero University Kano, PMB 3011 Kano State, Nigeria.
| | - King-Hwa Ling
- NeuroBiology & Genetics Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 UPM Serdang, Selangor, Malaysia.
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, 43400 UPM Serdang, Selangor, Malaysia.
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 UPM Serdang, Selangor, Malaysia.
| | - Jia-Yong Lam
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Hui-Yee Chee
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
72
|
Dissecting the Cell Entry Pathway of Baculovirus by Single-Particle Tracking and Quantitative Electron Microscopic Analysis. J Virol 2019; 93:JVI.00033-19. [PMID: 30760565 DOI: 10.1128/jvi.00033-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 02/07/2023] Open
Abstract
The budded virus of Autographa californica multiple nucleopolyhedrovirus (AcMNPV) infects insect cells through mainly clathrin-mediated endocytosis. However, the cell entry pathway of AcMNPV remains unclear. In this study, by using population-based analysis of single-virus tracking and electron microscopy, we investigated the internalization, fusion behavior, and endocytic trafficking of AcMNPV. AcMNPV internalization into host insect cells was facilitated by actin polymerization and dynamin. After incorporation into early endosomes, the AcMNPV envelope fused with the membranes of early endosome, allowing for nucleocapsid release into the cytoplasm. Microtubules were implicated in the bidirectional and long-range transport of virus-containing endosomes. In addition, microtubule depolymerization reduced the motility of virus-bearing early endosomes, impairing the progression of infection beyond enlarged early endosomes. These findings demonstrated that AcMNPV internalization was facilitated by actin polymerization in a dynamin-dependent manner, and nucleocapsid release occurred in early endosomes in a microtubule-dependent manner. This study provides mechanistic and kinetic insights into AcMNPV infection and enhance our understanding of the infection pathway of baculoviruses.IMPORTANCE Baculoviruses are used widely as environmentally benign pesticides, protein expression systems, and potential mammalian gene delivery vectors. Despite the significant application value, little is known about the cell entry and endocytic trafficking pathways of baculoviruses. In this study, we demonstrated that the alphabaculovirus AcMNPV exhibited actin- and microtubule-dependent transport for nucleocapsid release predominantly from within early endosomes. In contrast to AcMNPV transduction in mammalian cells, its infection in host insect cells is facilitated by actin polymerization for internalization and microtubules for endocytic trafficking within early endosomes, implying that AcMNPV exhibits cell type specificity in the requirement of the cytoskeleton network. In addition, experimental depolymerization of microtubules impaired the progression of infection beyond enlarged early endosomes. This is the first study that dissects the cell entry pathway of baculoviruses in host cells at the single-particle level, which advances our understanding of the early steps of baculovirus entry.
Collapse
|
73
|
Guo Y, Duan M, Wang X, Gao J, Guan Z, Zhang M. Early events in rabies virus infection—Attachment, entry, and intracellular trafficking. Virus Res 2019; 263:217-225. [DOI: 10.1016/j.virusres.2019.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/28/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
|
74
|
Okuda A, Tahara S, Hirose H, Takeuchi T, Nakase I, Ono A, Takehashi M, Tanaka S, Futaki S. Oligoarginine-Bearing Tandem Repeat Penetration-Accelerating Sequence Delivers Protein to Cytosol via Caveolae-Mediated Endocytosis. Biomacromolecules 2019; 20:1849-1859. [PMID: 30893557 DOI: 10.1021/acs.biomac.8b01299] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To facilitate the cytosolic delivery of larger molecules such as proteins, we developed a new cell-penetrating peptide sequence, named Pas2r12, consisting of a repeated Pas sequence (FFLIG-FFLIG) and d-dodeca-arginine (r12). This peptide significantly enhanced the cellular uptake and cytosolic release of enhanced green fluorescent protein and immunoglobulin G as cargos. We found that simply mixing Pas2r12 with cargos could generate cytosolic introducible forms. The cytosolic delivery of cargos by Pas2r12 was found to be an energy-requiring process, to rely on actin polymerization, and to be suppressed by caveolae-mediated endocytosis inhibitors (genistein and methyl-β-cyclodextrin) and small interfering RNA against caveolin-1. These results suggest that Pas2r12 enhances membrane penetration of cargos without the need for cross-linking and that caveolae-mediated endocytosis may be the route by which cytosolic delivery is enhanced.
Collapse
Affiliation(s)
- Akiko Okuda
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Shinya Tahara
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Hisaaki Hirose
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| | - Toshihide Takeuchi
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| | - Ikuhiko Nakase
- Graduate School of Science , Osaka Prefecture University , Naka-ku, Sakai , Osaka 599-8570 , Japan
| | - Atsushi Ono
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Masanori Takehashi
- Laboratory of Pathophysiology and Pharmacotherapeutics, Faculty of Pharmacy , Osaka Ohtani University , Tondabayashi , Osaka 584-8540 , Japan
| | - Seigo Tanaka
- Laboratory of Pathophysiology and Pharmacotherapeutics, Faculty of Pharmacy , Osaka Ohtani University , Tondabayashi , Osaka 584-8540 , Japan
| | - Shiroh Futaki
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| |
Collapse
|
75
|
Abstract
Recognition of cellular receptors allows emerging viruses to break species barriers and is an important determinant for their disease potential. Many virus receptors have complex tissue-specific interactomes, and preexisting protein-protein interactions may influence their function. Combining shotgun proteomics with a biochemical approach, we characterize the molecular composition of the functional receptor complexes used by the highly pathogenic Lassa virus (LASV) to invade susceptible human cells. We show that the specific composition of the receptor complexes affects productive entry of the virus, providing proof-of-concept. In uninfected cells, these functional receptor complexes undergo dynamic turnover involving an endocytic pathway that shares some characteristics with viral entry. However, steady-state receptor uptake and virus endocytosis critically differ in kinetics and underlying signaling, indicating that the pathogen can manipulate the receptor complex according to its needs. Our study highlights a remarkable complexity of LASV-receptor interaction and identifies possible targets for therapeutic antiviral intervention. Recognition of functional receptors by viruses is a key determinant for their host range, tissue tropism, and disease potential. The highly pathogenic Lassa virus (LASV) currently represents one of the most important emerging pathogens. The major cellular receptor for LASV in human cells is the ubiquitously expressed and evolutionary highly conserved extracellular matrix receptor dystroglycan (DG). In the host, DG interacts with many cellular proteins in a tissue-specific manner. The resulting distinct supramolecular complexes likely represent the functional units for viral entry, and preexisting protein-protein interactions may critically influence DG’s function in productive viral entry. Using an unbiased shotgun proteomic approach, we define the largely unknown molecular composition of DG complexes present in highly susceptible epithelial cells that represent important targets for LASV during viral transmission. We further show that the specific composition of cellular DG complexes can affect DG’s function in receptor-mediated endocytosis of the virus. Under steady-state conditions, epithelial DG complexes underwent rapid turnover via an endocytic pathway that shared some characteristics with DG-mediated LASV entry. However, compared to steady-state uptake of DG, LASV entry via DG occurred faster and critically depended on additional signaling by receptor tyrosine kinases and the downstream effector p21-activating kinase. In sum, we show that the specific molecular composition of DG complexes in susceptible cells is a determinant for productive virus entry and that the pathogen can manipulate the existing DG-linked endocytic pathway. This highlights another level of complexity of virus-receptor interaction and provides possible cellular targets for therapeutic antiviral intervention.
Collapse
|
76
|
Zhao T, Cui L, Yu X, Zhang Z, Shen X, Hua X. Entry of sapelovirus into IPEC-J2 cells is dependent on caveolae-mediated endocytosis. Virol J 2019; 16:37. [PMID: 30909932 PMCID: PMC6434631 DOI: 10.1186/s12985-019-1144-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/13/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Porcine sapelovirus (PSV), a species of the genus Sapelovirus within the family Picornaviridae, are a significant cause of enteritis, pneumonia, polioencephalomyelitis and reproductive disorders in pigs. However, the life cycle of PSV on the molecular level is largely unknown. METHODS Here, we used chemical inhibitors, RNA interference, and overexpression of dominant negative (DN) mutant plasmids to verify the roles of distinct endocytic pathways involved in PSV entry into porcine small intestinal epithelial cell line (IPEC-J2). RESULTS Our experiments indicated that PSV infection was inhibited when cells were pre-treated with NH4Cl or chloroquine. Inhibitors nystatin, methyl-β-cyclodextrin, dynasore and wortmannin dramatically reduced PSV entry efficiency, whereas the inhibitors chlorpromazine and EIPA had no effect. Furthermore, overexpression caveolin DN mutant and siRNA against caveolin also decreased virus titers and VP1 protein synthesis, whereas overexpression EPS15 DN mutant and siRNA against EPS15 did not reduce virus infection. CONCLUSIONS Our findings suggest that PSV entry into IPEC-J2 cells depends on caveolae/lipid raft mediated-endocytosis, that is pH-dependent and requires dynamin and PI3K but is independent of clathrin and macropinocytosis.
Collapse
Affiliation(s)
- Tingting Zhao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Li Cui
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Xiangqian Yu
- Shanghai Pudong New Area Center for Animal Disease Control and Prevention, Shanghai, 200136, China
| | - Zhonghai Zhang
- Shanghai Pudong New Area Center for Animal Disease Control and Prevention, Shanghai, 200136, China
| | - Xiaojuan Shen
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China
| | - Xiuguo Hua
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, China.
| |
Collapse
|
77
|
Infectious Entry of Merkel Cell Polyomavirus. J Virol 2019; 93:JVI.02004-18. [PMID: 30626687 DOI: 10.1128/jvi.02004-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/23/2018] [Indexed: 12/17/2022] Open
Abstract
Merkel cell polyomavirus (MCPyV) is a small, nonenveloped tumor virus associated with an aggressive form of skin cancer, Merkel cell carcinoma (MCC). MCPyV infections are highly prevalent in the human population, with MCPyV virions being continuously shed from human skin. However, the precise host cell tropism(s) of MCPyV remains unclear: MCPyV is able to replicate within a subset of dermal fibroblasts, but MCPyV DNA has also been detected in a variety of other tissues. However, MCPyV appears different from other polyomaviruses, as it requires sulfated polysaccharides, such as heparan sulfates and/or chondroitin sulfates, for initial attachment. Like other polyomaviruses, MCPyV engages sialic acid as a (co)receptor. To explore the infectious entry process of MCPyV, we analyzed the cell biological determinants of MCPyV entry into A549 cells, a highly transducible lung carcinoma cell line, in comparison to well-studied simian virus 40 and a number of other viruses. Our results indicate that MCPyV enters cells via caveolar/lipid raft-mediated endocytosis but not macropinocytosis, clathrin-mediated endocytosis, or glycosphingolipid-enriched carriers. The viruses were internalized in small endocytic pits that led the virus to endosomes and from there to the endoplasmic reticulum (ER). Similar to other polyomaviruses, trafficking required microtubular transport, acidification of endosomes, and a functional redox environment. To our surprise, the virus was found to acquire a membrane envelope within endosomes, a phenomenon not reported for other viruses. Only minor amounts of viruses reached the ER, while the majority was retained in endosomal compartments, suggesting that endosome-to-ER trafficking is a bottleneck during infectious entry.IMPORTANCE MCPyV is the first polyomavirus directly implicated in the development of an aggressive human cancer, Merkel cell carcinoma (MCC). Although MCPyV is constantly shed from healthy skin, the MCC incidence increases among aging and immunocompromised individuals. To date, the events connecting initial MCPyV infection and subsequent transformation still remain elusive. MCPyV differs from other known polyomaviruses concerning its cell tropism, entry receptor requirements, and infection kinetics. In this study, we examined the cellular requirements for endocytic entry as well as the subcellular localization of incoming virus particles. A thorough understanding of the determinants of the infectious entry pathway and the specific biological niche will benefit prevention of virus-derived cancers such as MCC.
Collapse
|
78
|
Identification of Clotrimazole Derivatives as Specific Inhibitors of Arenavirus Fusion. J Virol 2019; 93:JVI.01744-18. [PMID: 30626681 DOI: 10.1128/jvi.01744-18] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
Arenaviruses are a large family of emerging enveloped negative-strand RNA viruses that include several causative agents of viral hemorrhagic fevers. For cell entry, human-pathogenic arenaviruses use different cellular receptors and endocytic pathways that converge at the level of acidified late endosomes, where the viral envelope glycoprotein mediates membrane fusion. Inhibitors of arenavirus entry hold promise for therapeutic antiviral intervention and the identification of "druggable" targets is of high priority. Using a recombinant vesicular stomatitis virus pseudotype platform, we identified the clotrimazole-derivative TRAM-34, a highly selective antagonist of the calcium-activated potassium channel KCa3.1, as a specific entry inhibitor for arenaviruses. TRAM-34 specifically blocked entry of most arenaviruses, including hemorrhagic fever viruses, but not Lassa virus and other enveloped viruses. Anti-arenaviral activity was likewise observed with the parental compound clotrimazole and the derivative senicapoc, whereas structurally unrelated KCa3.1 inhibitors showed no antiviral effect. Deletion of KCa3.1 by CRISPR/Cas9 technology did not affect the antiarenaviral effect of TRAM-34, indicating that the observed antiviral effect of clotrimazoles was independent of the known pharmacological target. The drug affected neither virus-cell attachment, nor endocytosis, suggesting an effect on later entry steps. Employing a quantitative cell-cell fusion assay that bypasses endocytosis, we demonstrate that TRAM-34 specifically inhibits arenavirus-mediated membrane fusion. In sum, we uncover a novel antiarenaviral action of clotrimazoles that currently undergo in vivo evaluation in the context of other human diseases. Their favorable in vivo toxicity profiles and stability opens the possibility to repurpose clotrimazole derivatives for therapeutic intervention against human-pathogenic arenaviruses.IMPORTANCE Emerging human-pathogenic arenaviruses are causative agents of severe hemorrhagic fevers with high mortality and represent serious public health problems. The current lack of a licensed vaccine and the limited treatment options makes the development of novel antiarenaviral therapeutics an urgent need. Using a recombinant pseudotype platform, we uncovered that clotrimazole drugs, in particular TRAM-34, specifically inhibit cell entry of a range of arenaviruses, including important emerging human pathogens, with the exception of Lassa virus. The antiviral effect was independent of the known pharmacological drug target and involved inhibition of the unusual membrane fusion mechanism of arenaviruses. TRAM-34 and its derivatives currently undergo evaluation against a number of human diseases and show favorable toxicity profiles and high stability in vivo Our study provides the basis for further evaluation of clotrimazole derivatives as antiviral drug candidates. Their advanced stage of drug development will facilitate repurposing for therapeutic intervention against human-pathogenic arenaviruses.
Collapse
|
79
|
Lin HJ, Jiang ZP, Lo HR, Feng CL, Chen CJ, Yang CY, Huang MZ, Wu HY, Chen YA, Chen Y, Chiu CH, Lai CH. Coalescence of RAGE in Lipid Rafts in Response to Cytolethal Distending Toxin-Induced Inflammation. Front Immunol 2019; 10:109. [PMID: 30863392 PMCID: PMC6399302 DOI: 10.3389/fimmu.2019.00109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) interacts with various molecules in the cell membrane to induce an inflammatory response. The cytolethal distending toxin (CDT) produced by Campylobacter jejuni contains three subunits: CdtA, CdtB, and CdtC. Amongst, CdtA and CdtC interact with membrane lipid rafts, by which CdtB enters the nucleus to induce pathogenesis. In this study, we first explored the relationships between RAGE, lipid rafts, and inflammation in gastrointestinal epithelial cells exposed to CDT. Our results showed that CDT activated the expression of RAGE and high mobility group box 1 (HMGB1), followed by the recruitment of RAGE into lipid rafts. In contrast, RAGE antagonist inhibited CDT-induced inflammation via the RAGE-HMGB1 axis. Disruption of lipid rafts decreased CDT-induced downstream signaling, which in turn attenuated the inflammatory response. Furthermore, in vivo studies revealed severe inflammation and upregulation of RAGE and IL-1β in the intestinal tissues of CDT-treated mice. These results demonstrate that mobilization of RAGE to lipid rafts plays a crucial role in CDT-induced inflammation.
Collapse
Affiliation(s)
- Hwai-Jeng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho Hospital, New Taipei, Taiwan
| | - Zhi-Pei Jiang
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Colon and Rectal Surgery, Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Horng-Ren Lo
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Chun-Lung Feng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Jung Chen
- Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chia-Yu Yang
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Colon and Rectal Surgery, Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Mei-Zi Huang
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-An Chen
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu Chen
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
80
|
Nathan L, Daniel S. Single Virion Tracking Microscopy for the Study of Virus Entry Processes in Live Cells and Biomimetic Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1215:13-43. [PMID: 31317494 PMCID: PMC7122913 DOI: 10.1007/978-3-030-14741-9_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The most widely-used assays for studying viral entry, including infectivity, cofloatation, and cell-cell fusion assays, yield functional information but provide low resolution of individual entry steps. Structural characterization provides high-resolution conformational information, but on its own is unable to address the functional significance of these conformations. Single virion tracking microscopy techniques provide more detail on the intermediate entry steps than infection assays and more functional information than structural methods, bridging the gap between these methods. In addition, single virion approaches also provide dynamic information about the kinetics of entry processes. This chapter reviews single virion tracking techniques and describes how they can be applied to study specific virus entry steps. These techniques provide information complementary to traditional ensemble approaches. Single virion techniques may either probe virion behavior in live cells or in biomimetic platforms. Synthesizing information from ensemble, structural, and single virion techniques ultimately yields a more complete understanding of the viral entry process than can be achieved by any single method alone.
Collapse
Affiliation(s)
- Lakshmi Nathan
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
81
|
Cellular Uptake Mechanisms and Detection of Nanoparticle Uptake by Advanced Imaging Methods. BIOLOGICAL RESPONSES TO NANOSCALE PARTICLES 2019. [DOI: 10.1007/978-3-030-12461-8_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
82
|
Autographa Californica Multiple Nucleopolyhedrovirus Enters Host Cells via Clathrin-Mediated Endocytosis and Direct Fusion with the Plasma Membrane. Viruses 2018; 10:v10110632. [PMID: 30441758 PMCID: PMC6266293 DOI: 10.3390/v10110632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 01/08/2023] Open
Abstract
The cell entry mechanism of Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is not fully understood. Previous studies showed that AcMNPV entered host cells primarily through clathrin-mediated endocytosis, and could efficiently infect cells via fusion with the plasma membrane after a low-pH trigger. However, whether AcMNPV enters cells via these two pathways simultaneously, and the exact manner in which AcMNPV particles are internalized into cells remains unclear. In this study, using single-virus tracking, we observed that AcMNPV particles were first captured by pre-existing clathrin-coated pits (CCP), and were then delivered to early endosomes. Population-based analysis of single-virus tracking and quantitative electron microscopy demonstrated that the majority of particles were captured by CCPs and internalized via invagination. In contrast, a minority of virus particles were not delivered to CCPs, and were internalized through direct fusion with the plasma membrane without invagination. Quantitative electron microscopy also showed that, while inhibition of CCP assembly significantly impaired viral internalization, inhibition of endosomal acidification blocked virus particles out of vesicles. Collectively, these findings demonstrated that approximately 90% of AcMNPV particles entered cells through clathrin-mediated endocytosis and 10% entered via direct fusion with the plasma membrane. This study will lead toward a better understanding of AcMNPV infection.
Collapse
|
83
|
Reinholz J, Landfester K, Mailänder V. The challenges of oral drug delivery via nanocarriers. Drug Deliv 2018; 25:1694-1705. [PMID: 30394120 PMCID: PMC6225504 DOI: 10.1080/10717544.2018.1501119] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 01/11/2023] Open
Abstract
The oral application of pharmaceuticals is unarguably the most convenient method of application. Especially for protein- or peptide-based drugs, however, the effectiveness is significantly reduced due to enzymatic digestion in the stomach as well as a poor bioavailability in the small intestine. For these difficult formulations, the encapsulation into nanocarriers would protect the sensitive drug and thus could considerably improve the efficiency of oral drug delivery. In the last years, many candidate biodegradable nanomaterials for such carrier systems have been published. However, before the cargo can be released, the nanocarrier needs to cross multiple barriers of the human body, including a layer of intestinal mucus and epithelial as well as endothelial cells. For overcoming these cellular barriers, transcytosis is favored over a paracellular transport for most nanomaterials as paracellular transport routes lack selectivity of transported molecules once opened up. The exact mechanisms behind the transcellular translocations are up to now still not completely understood. For the vast majority of nanocarriers, the rate of transcellular transport is not sufficient to realize their application in oral drug delivery. Especially trafficking into the endolysosomal pathway often marks a key problem. In this review, we focus on the molecular mechanisms of overcoming cellular barriers, especially transcytosis, and highlight difficulties of oral drug delivery via nanocarriers.
Collapse
Affiliation(s)
- Jonas Reinholz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Volker Mailänder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| |
Collapse
|
84
|
Sharma KK, Marzinek JK, Tantirimudalige SN, Bond PJ, Wohland T. Single-molecule studies of flavivirus envelope dynamics: Experiment and computation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 143:38-51. [PMID: 30223001 DOI: 10.1016/j.pbiomolbio.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022]
Abstract
Flaviviruses are simple enveloped viruses exhibiting complex structural and functional heterogeneities. Decades of research have provided crucial basic insights, antiviral medication and moderately successful gene therapy trials. The most infectious particle is, however, not always the most abundant one in a population, questioning the utility of classic ensemble-averaging virology approaches. Indeed, viral replication is often not particularly efficient, prone to errors or containing parallel routes. Here, we review different single-molecule sensitive fluorescence methods that are employed to investigate flaviviruses. In particular, we review how (i) time-resolved Förster resonance energy transfer (trFRET) was applied to probe dengue envelope conformations; (ii) FRET-fluorescence correlation spectroscopy to investigate dengue envelope intrinsic dynamics and (iii) single particle tracking to follow the path of dengue viruses in cells. We also discuss how such methods may be supported by molecular dynamics (MD) simulations over a range of spatio-temporal scales, to provide complementary data on the structure and dynamics of flaviviral systems. We describe recent improvements in multiscale MD approaches that allowed the simulation of dengue particle envelopes in near-atomic resolution. We hope this review is an incentive for setting up and applying similar single-molecule studies and combine them with MD simulations to investigate structural dynamics of entire flavivirus particles over the nanosecond-to-millisecond time-scale and follow viruses during infection in cells over milliseconds to minutes.
Collapse
Affiliation(s)
- Kamal Kant Sharma
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Jan K Marzinek
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Sarala Neomi Tantirimudalige
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Peter J Bond
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore.
| | - Thorsten Wohland
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Department of Chemistry, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Centre for Bioimaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117557, Singapore.
| |
Collapse
|
85
|
Sánchez EE, González R, Lucena S, García S, Finol HJ, Suntravat M, Girón ME, Fernández I, Rodríguez-Acosta A. Crotamine-like from Southern Pacific rattlesnake (Crotalus oreganus helleri) Venom acts on human leukemia (K-562) cell lines and produces ultrastructural changes on mice adrenal gland. Ultrastruct Pathol 2018; 42:116-123. [PMID: 29466088 DOI: 10.1080/01913123.2017.1422827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Crotamine is a cationic, non-enzymatic, protein integrating a minor family of myotoxins, composed of 42 amino acid residues, described in Viperidae and Crotalidae snake's families that has been used in neuroscience research, drug progressing and molecular diversity reports. Crotamine-like protein (CLP) from C.o.helleri venom was isolated in fraction 5 from 7 peaks obtained by sulfopropyl waters protein pak cationic exchange column. In tricine-SDS-PAGE under non-reduced conditions this CLP showed a single band of ~8 kDa molecular weight. CLP induced toxicity of K-562 cells with a CC50 of 11.09 µM. In mice adrenal gland after 24 h of CLP injection, cortical cells exhibited swollen mitochondria with scarce tubular cristae, some elements of smooth and rough endoplasmic reticula, widened nuclear envelope, slightly osmiophilic lipid droplets, and autophagic vacuoles. In some areas cortical cells plasma membrane and endothelial walls disappeared, which indicated a necrosis process. In other areas, endothelial cell cytoplasm did not present the normal caveolae and pinocytotic vesicles. To our knowledge, this is the first report on mice adrenal gland damages, caused by the injection of CLP from rattlesnakes. Our results propose that adrenal cortex lesions may be significant in the envenoming etiopathogenesis by CLP.
Collapse
Affiliation(s)
- Elda E Sánchez
- a National Natural Toxins Research Center (NNTRC) , Texas A&M University-Kingsville , Kingsville , TX , USA.,d Department of Chemistry , Texas A&M University-Kingsville , Kingsville , TX , USA
| | - Roschman González
- b Centro de Microscopia Electronica , Facultad de Ciencias de la Universidad Central de Venezuela , Caracas , Venezuela
| | - Sara Lucena
- a National Natural Toxins Research Center (NNTRC) , Texas A&M University-Kingsville , Kingsville , TX , USA
| | - Stefanie García
- b Centro de Microscopia Electronica , Facultad de Ciencias de la Universidad Central de Venezuela , Caracas , Venezuela
| | - Héctor J Finol
- b Centro de Microscopia Electronica , Facultad de Ciencias de la Universidad Central de Venezuela , Caracas , Venezuela
| | - Montamas Suntravat
- a National Natural Toxins Research Center (NNTRC) , Texas A&M University-Kingsville , Kingsville , TX , USA
| | - María E Girón
- c Laboratorio de Inmunoquímica y Ultraestructura , Instituto Anatómico de la Universidad Central de Venezuela , Caracas , Venezuela
| | - Irma Fernández
- c Laboratorio de Inmunoquímica y Ultraestructura , Instituto Anatómico de la Universidad Central de Venezuela , Caracas , Venezuela
| | - Alexis Rodríguez-Acosta
- c Laboratorio de Inmunoquímica y Ultraestructura , Instituto Anatómico de la Universidad Central de Venezuela , Caracas , Venezuela
| |
Collapse
|
86
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
87
|
Toscano MG, de Haan P. How Simian Virus 40 Hijacks the Intracellular Protein Trafficking Pathway to Its Own Benefit … and Ours. Front Immunol 2018; 9:1160. [PMID: 29892296 PMCID: PMC5985306 DOI: 10.3389/fimmu.2018.01160] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/09/2018] [Indexed: 12/29/2022] Open
Abstract
Viruses efficiently transfer and express their genes in host cells and evolve to evade the host's defense responses. These properties render them highly attractive for use as gene delivery vectors in vaccines, gene, and immunotherapies. Among the viruses used as gene delivery vectors, the macaque polyomavirus Simian Virus 40 (SV40) is unique in its capacity to evade intracellular antiviral defense responses upon cell entry. We here describe the unique way by which SV40 particles deliver their genomes in the nucleus of permissive cells and how they prevent presentation of viral antigens to the host's immune system. The non-immunogenicity in its natural host is not only of benefit to the virus but also to us in developing effective SV40 vector-based treatments for today's major human diseases.
Collapse
|
88
|
Owczarek K, Szczepanski A, Milewska A, Baster Z, Rajfur Z, Sarna M, Pyrc K. Early events during human coronavirus OC43 entry to the cell. Sci Rep 2018; 8:7124. [PMID: 29740099 PMCID: PMC5940804 DOI: 10.1038/s41598-018-25640-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/25/2018] [Indexed: 12/23/2022] Open
Abstract
The Coronaviridae family clusters a number of large RNA viruses, which share several structural and functional features. However, members of this family recognize different cellular receptors and exploit different entry routes, what affects their species specificity and virulence. The aim of this study was to determine how human coronavirus OC43 enters the susceptible cell. Using confocal microscopy and molecular biology tools we visualized early events during infection. We found that the virus employs caveolin-1 dependent endocytosis for the entry and the scission of virus-containing vesicles from the cell surface is dynamin-dependent. Furthermore, the vesicle internalization process requires actin cytoskeleton rearrangements. With our research we strove to broaden the understanding of the infection process, which in future may be beneficial for the development of a potential therapeutics.
Collapse
Affiliation(s)
- Katarzyna Owczarek
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
| | - Artur Szczepanski
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
| | - Aleksandra Milewska
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
| | - Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Michal Sarna
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Krzysztof Pyrc
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland.
| |
Collapse
|
89
|
Picco A, Kukulski W, Manenschijn HE, Specht T, Briggs JAG, Kaksonen M. The contributions of the actin machinery to endocytic membrane bending and vesicle formation. Mol Biol Cell 2018; 29:1346-1358. [PMID: 29851558 PMCID: PMC5994895 DOI: 10.1091/mbc.e17-11-0688] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Branched and cross-linked actin networks mediate cellular processes that move and shape membranes. To understand how actin contributes during the different stages of endocytic membrane reshaping, we analyzed deletion mutants of yeast actin network components using a hybrid imaging approach that combines live imaging with correlative microscopy. We could thus temporally dissect the effects of different actin network perturbations, revealing distinct stages of actin-based membrane reshaping. Our data show that initiation of membrane bending requires the actin network to be physically linked to the plasma membrane and to be optimally cross-linked. Once initiated, the membrane invagination process is driven by nucleation and polymerization of new actin filaments, independent of the degree of cross-linking and unaffected by a surplus of actin network components. A key transition occurs 2 s before scission, when the filament nucleation rate drops. From that time point on, invagination growth and vesicle scission are driven by an expansion of the actin network without a proportional increase of net actin amounts. The expansion is sensitive to the amount of filamentous actin and its cross-linking. Our results suggest that the mechanism by which actin reshapes the membrane changes during the progress of endocytosis, possibly adapting to varying force requirements.
Collapse
Affiliation(s)
- Andrea Picco
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Wanda Kukulski
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Hetty E Manenschijn
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Tanja Specht
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - John A G Briggs
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Marko Kaksonen
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
90
|
Imaging, Tracking and Computational Analyses of Virus Entry and Egress with the Cytoskeleton. Viruses 2018; 10:v10040166. [PMID: 29614729 PMCID: PMC5923460 DOI: 10.3390/v10040166] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Viruses have a dual nature: particles are “passive substances” lacking chemical energy transformation, whereas infected cells are “active substances” turning-over energy. How passive viral substances convert to active substances, comprising viral replication and assembly compartments has been of intense interest to virologists, cell and molecular biologists and immunologists. Infection starts with virus entry into a susceptible cell and delivers the viral genome to the replication site. This is a multi-step process, and involves the cytoskeleton and associated motor proteins. Likewise, the egress of progeny virus particles from the replication site to the extracellular space is enhanced by the cytoskeleton and associated motor proteins. This overcomes the limitation of thermal diffusion, and transports virions and virion components, often in association with cellular organelles. This review explores how the analysis of viral trajectories informs about mechanisms of infection. We discuss the methodology enabling researchers to visualize single virions in cells by fluorescence imaging and tracking. Virus visualization and tracking are increasingly enhanced by computational analyses of virus trajectories as well as in silico modeling. Combined approaches reveal previously unrecognized features of virus-infected cells. Using select examples of complementary methodology, we highlight the role of actin filaments and microtubules, and their associated motors in virus infections. In-depth studies of single virion dynamics at high temporal and spatial resolutions thereby provide deep insight into virus infection processes, and are a basis for uncovering underlying mechanisms of how cells function.
Collapse
|
91
|
Huang B, Yan S, Xiao L, Ji R, Yang L, Miao AJ, Wang P. Label-Free Imaging of Nanoparticle Uptake Competition in Single Cells by Hyperspectral Stimulated Raman Scattering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:1703246. [PMID: 29283225 DOI: 10.1002/smll.201703246] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/14/2017] [Indexed: 06/07/2023]
Abstract
Imaging and quantification of nanoparticles in single cells in their most natural condition are expected to facilitate the biotechnological applications of nanoparticles and allow for better assessment of their biosafety risks. However, current imaging modalities either require tedious sample preparation or only apply to nanoparticles with specific physicochemical characteristics. Here, the emerging hyperspectral stimulated Raman scattering (SRS) microscopy, as a label-free and nondestructive imaging method, is used for the first time to investigate the subcellular distribution of nanoparticles in the protozoan Tetrahymena thermophila. The two frequently studied nanoparticles, polyacrylate-coated α-Fe2 O3 and TiO2 , are found to have different subcellular distribution pattern as a result of their dissimilar uptake routes. Significant uptake competition between these two types of nanoparticles is further discovered, which should be paid attention to in future bioapplications of nanoparticles. Overall, this study illustrates the great promise of hyperspectral SRS as an analytical imaging tool in nanobiotechnology and nanotoxicology.
Collapse
Affiliation(s)
- Bin Huang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu Province, China
| | - Shuai Yan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Lin Xiao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu Province, China
| | - Rong Ji
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu Province, China
| | - Liuyan Yang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu Province, China
| | - Ai-Jun Miao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu Province, China
| | - Ping Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| |
Collapse
|
92
|
Dixit S, Sahu R, Verma R, Duncan S, Giambartolomei GH, Singh SR, Dennis VA. Caveolin-mediated endocytosis of the Chlamydia M278 outer membrane peptide encapsulated in poly(lactic acid)-Poly(ethylene glycol) nanoparticles by mouse primary dendritic cells enhances specific immune effectors mediated by MHC class II and CD4 + T cells. Biomaterials 2018; 159:130-145. [PMID: 29324305 PMCID: PMC5801148 DOI: 10.1016/j.biomaterials.2017.12.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 12/18/2022]
Abstract
We previously developed a Chlamydia trachomatis nanovaccine (PPM) by encapsulating a chlamydial M278 peptide within poly(lactic acid)-poly(ethylene glycol) biodegradable nanoparticles that immunopotentiated Chlamydia-specific immune effector responses in mice. Herein, we investigated the mechanistic interactions of PPM with mouse bone marrow-derived dendritic cells (DCs) for its uptake, trafficking, and T cell activation. Our results reveal that PPM triggered enhanced expression of effector cytokines and chemokines, surface activation markers (Cd1d2, Fcgr1), pathogen-sensing receptors (TLR2, Nod1), co-stimulatory (CD40, CD80, CD86) and MHC class I and II molecules. Co-culturing of PPM-primed DCs with T cells from C. muridarum vaccinated mice yielded an increase in Chlamydia-specific immune effector responses including CD3+ lymphoproliferation, CD3+CD4+ IFN-γ-secreting cells along with CD3+CD4+ memory (CD44high and CD62Lhigh) and effector (CD44high and CD62Llow) phenotypes. Intracellular trafficking analyses revealed an intense expression and colocalization of PPM predominantly in endosomes. PPM also upregulated the transcriptional and protein expression of the endocytic mediator, caveolin-1 in DCs. More importantly, the specific inhibition of caveolin-1 led to decreased expression of PPM-induced cytokines and co-stimulatory molecules. Our investigation shows that PPM provided enhancement of uptake, probably by exploiting the caveolin-mediated endocytosis pathway, endosomal processing, and MHC II presentation to immunopotentiate Chlamydia-specific immune effector responses mediated by CD4+ T cells.
Collapse
Affiliation(s)
- Saurabh Dixit
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA
| | - Rajnish Sahu
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA
| | - Richa Verma
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA
| | - Skyla Duncan
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Shree R Singh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
93
|
Edlich A, Volz P, Brodwolf R, Unbehauen M, Mundhenk L, Gruber AD, Hedtrich S, Haag R, Alexiev U, Kleuser B. Crosstalk between core-multishell nanocarriers for cutaneous drug delivery and antigen-presenting cells of the skin. Biomaterials 2018; 162:60-70. [PMID: 29438881 DOI: 10.1016/j.biomaterials.2018.01.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/24/2018] [Accepted: 01/31/2018] [Indexed: 01/15/2023]
Abstract
Owing their unique chemical and physical properties core-multishell (CMS) nanocarriers are thought to underlie their exploitable biomedical use for a topical treatment of skin diseases. This highlights the need to consider not only the efficacy of CMS nanocarriers but also the potentially unpredictable and adverse consequences of their exposure thereto. As CMS nanocarriers are able to penetrate into viable layers of normal and stripped human skin ex vivo as well as in in vitro skin disease models the understanding of nanoparticle crosstalk with components of the immune system requires thorough investigation. Our studies highlight the biocompatible properties of CMS nanocarriers on Langerhans cells of the skin as they did neither induce cytotoxicity and genotoxicity nor cause reactive oxygen species (ROS) or an immunological response. Nevertheless, CMS nanocarriers were efficiently taken up by Langerhans cells via divergent endocytic pathways. Bioimaging of CMS nanocarriers by fluorescence lifetime imaging microscopy (FLIM) and flow cytometry indicated not only a localization within the lysosomes but also an energy-dependent exocytosis of unmodified CMS nanocarriers into the extracellular environment.
Collapse
Affiliation(s)
- Alexander Edlich
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Pierre Volz
- Institute of Experimental Physics, Freie Universität Berlin, Berlin, Germany
| | - Robert Brodwolf
- Institute of Experimental Physics, Freie Universität Berlin, Berlin, Germany
| | - Michael Unbehauen
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Sarah Hedtrich
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Ulrike Alexiev
- Institute of Experimental Physics, Freie Universität Berlin, Berlin, Germany.
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany.
| |
Collapse
|
94
|
Kou L, Bhutia YD, Yao Q, He Z, Sun J, Ganapathy V. Transporter-Guided Delivery of Nanoparticles to Improve Drug Permeation across Cellular Barriers and Drug Exposure to Selective Cell Types. Front Pharmacol 2018; 9:27. [PMID: 29434548 PMCID: PMC5791163 DOI: 10.3389/fphar.2018.00027] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 12/17/2022] Open
Abstract
Targeted nano-drug delivery systems conjugated with specific ligands to target selective cell-surface receptors or transporters could enhance the efficacy of drug delivery and therapy. Transporters are expressed differentially on the cell-surface of different cell types, and also specific transporters are expressed at higher than normal levels in selective cell types under pathological conditions. They also play a key role in intestinal absorption, delivery via non-oral routes (e.g., pulmonary route and nasal route), and transfer across biological barriers (e.g., blood–brain barrier and blood–retinal barrier. As such, the cell-surface transporters represent ideal targets for nano-drug delivery systems to facilitate drug delivery to selective cell types under normal or pathological conditions and also to avoid off-target adverse side effects of the drugs. There is increasing evidence in recent years supporting the utility of cell-surface transporters in the field of nano-drug delivery to increase oral bioavailability, to improve transfer across the blood–brain barrier, and to enhance delivery of therapeutics in a cell-type selective manner in disease states. Here we provide a comprehensive review of recent advancements in this interesting and important area. We also highlight certain key aspects that need to be taken into account for optimal development of transporter-assisted nano-drug delivery systems.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yangzom D Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Qing Yao
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhonggui He
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Jin Sun
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
95
|
Abstract
Host cell entry is the first and most fundamental step of every virus infection and represents a major barrier for zoonotic transmission and viral emergence. Targeting viral entry appears further as a promising strategy for therapeutic intervention. Several cellular receptors have been identified for Lassa virus, including dystroglycan, TAM receptor tyrosine kinases, and C-type lectins. Upon receptor binding, LASV enters the host cell via a largely unknown clathrin- and dynamin-independent endocytotic pathway that delivers the virus to late endosomes, where fusion occurs after engagement of a second, intracellular receptor, the late endosomal/lysosomal resident protein LAMP1. Here, we describe a series of experimental approaches to investigate LASV cell entry and to test candidate inhibitors for their action at this early and decisive step of infection.
Collapse
Affiliation(s)
- Antonella Pasquato
- Institute of Microbiology, University Hospital Center, University of Lausanne, Rue du Bugnon 48, CH-1011, Lausanne, Switzerland
| | - Antonio Herrador Fernandez
- Institute of Microbiology, University Hospital Center, University of Lausanne, Rue du Bugnon 48, CH-1011, Lausanne, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, University Hospital Center, University of Lausanne, Rue du Bugnon 48, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
96
|
Hairpin probe for sequence-specific recognition of double-stranded DNA on simian virus 40. Chem Res Chin Univ 2017. [DOI: 10.1007/s40242-017-7152-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
97
|
Burton RAB, Rog-Zielinska EA, Corbett AD, Peyronnet R, Bodi I, Fink M, Sheldon J, Hoenger A, Calaghan SC, Bub G, Kohl P. Caveolae in Rabbit Ventricular Myocytes: Distribution and Dynamic Diminution after Cell Isolation. Biophys J 2017; 113:1047-1059. [PMID: 28877488 PMCID: PMC5653872 DOI: 10.1016/j.bpj.2017.07.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/16/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022] Open
Abstract
Caveolae are signal transduction centers, yet their subcellular distribution and preservation in cardiac myocytes after cell isolation are not well documented. Here, we quantify caveolae located within 100 nm of the outer cell surface membrane in rabbit single-ventricular cardiomyocytes over 8 h post-isolation and relate this to the presence of caveolae in intact tissue. Hearts from New Zealand white rabbits were either chemically fixed by coronary perfusion or enzymatically digested to isolate ventricular myocytes, which were subsequently fixed at 0, 3, and 8 h post-isolation. In live cells, the patch-clamp technique was used to measure whole-cell plasma membrane capacitance, and in fixed cells, caveolae were quantified by transmission electron microscopy. Changes in cell-surface topology were assessed using scanning electron microscopy. In fixed ventricular myocardium, dual-axis electron tomography was used for three-dimensional reconstruction and analysis of caveolae in situ. The presence and distribution of surface-sarcolemmal caveolae in freshly isolated cells matches that of intact myocardium. With time, the number of surface-sarcolemmal caveolae decreases in isolated cardiomyocytes. This is associated with a gradual increase in whole-cell membrane capacitance. Concurrently, there is a significant increase in area, diameter, and circularity of sub-sarcolemmal mitochondria, indicative of swelling. In addition, electron tomography data from intact heart illustrate the regular presence of caveolae not only at the surface sarcolemma, but also on transverse-tubular membranes in ventricular myocardium. Thus, caveolae are dynamic structures, present both at surface-sarcolemmal and transverse-tubular membranes. After cell isolation, the number of surface-sarcolemmal caveolae decreases significantly within a time frame relevant for single-cell research. The concurrent increase in cell capacitance suggests that membrane incorporation of surface-sarcolemmal caveolae underlies this, but internalization and/or micro-vesicle loss to the extracellular space may also contribute. Given that much of the research into cardiac caveolae-dependent signaling utilizes isolated cells, and since caveolae-dependent pathways matter for a wide range of other study targets, analysis of isolated cell data should take the time post-isolation into account.
Collapse
Affiliation(s)
- Rebecca A B Burton
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Eva A Rog-Zielinska
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ilona Bodi
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Fink
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Judith Sheldon
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Andreas Hoenger
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Sarah C Calaghan
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Gil Bub
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
98
|
Sun J, Liu Y, Ge M, Zhou G, Sun W, Liu D, Liang XJ, Zhang J. A Distinct Endocytic Mechanism of Functionalized-Silica Nanoparticles in Breast Cancer Stem Cells. Sci Rep 2017; 7:16236. [PMID: 29176652 PMCID: PMC5701218 DOI: 10.1038/s41598-017-16591-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/06/2017] [Indexed: 01/01/2023] Open
Abstract
Nanoparticles provide new fields for life medical science application, including targeted-drug delivery and cancer treatment. To maximize the delivery efficiency of nanoparticle, one must understand the uptake mechanism of nanoparticle in cells, which may determine their ultimate fate and localization in cells. Recently, the proposed-cancer stem cell (CSC) theory has been attracted great attention and regarded as new targets for the new nanodrug developmet and cancer therapies. The interaction between nanoparticles and cancer cells has been extensively studied, but the uptake mechanism of nanoparticles in CSCs has received little attention. Here, we use the pharmacological inhibitors of major endocytic pathways to study the silica nanoparticle (SiNP) uptake mechanisms in the human breast adenocarcinoma cell line (MCF-7) and MCF-7-derived breast cancer stem cells (BCSCs). The results demonstrate that the uptake of SiNPs, particularly amino-functionalized SiNPs, in MCF-7 cells is strongly affected by the actin depolymerization, whereas BCSCs more strongly inhibit the amino-functionalized SiNP uptake after the scavenger receptor disruption. These findings indicate a distinct endocytic mechanism of functionalized SiNPs in BCSCs, which is significant for designing ideal nanosized drug delivery systems and improving the selectivity for CSC-targeted therapy.
Collapse
Affiliation(s)
- Jiadong Sun
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China.,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China.,CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, People's Republic of China
| | - Yajing Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China.,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Min Ge
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China.,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Guoqiang Zhou
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China.,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Wentong Sun
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China.,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Dandan Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China. .,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China.
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, People's Republic of China.
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, People's Republic of China. .,College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China.
| |
Collapse
|
99
|
Costa Verdera H, Gitz-Francois JJ, Schiffelers RM, Vader P. Cellular uptake of extracellular vesicles is mediated by clathrin-independent endocytosis and macropinocytosis. J Control Release 2017; 266:100-108. [PMID: 28919558 DOI: 10.1016/j.jconrel.2017.09.019] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/11/2022]
Abstract
Recent evidence has established that extracellular vesicles (EVs), including exosomes and microvesicles, form an endogenous transport system through which biomolecules, including proteins and RNA, are exchanged between cells. This endows EVs with immense potential for drug delivery and regenerative medicine applications. Understanding the biology underlying EV-based intercellular transfer of cargo is of great importance for the development of EV-based therapeutics. Here, we sought to characterize the cellular mechanisms involved in EV uptake. Internalization of fluorescently-labeled EVs was evaluated in HeLa cells, in 2D (monolayer) cell culture as well as 3D spheroids. Uptake was assessed using flow cytometry and confocal microscopy, using chemical as well as RNA interference-based inhibition of key proteins involved in individual endocytic pathways. Experiments with chemical inhibitors revealed that EV uptake depends on cholesterol and tyrosine kinase activity, which are implicated in clathrin-independent endocytosis, and on Na+/H+ exchange and phosphoinositide 3-kinase activity, which are important for macropinocytosis. Furthermore, EV internalization was inhibited by siRNA-mediated knockdown of caveolin-1, flotillin-1, RhoA, Rac1 and PAK1, but not clathrin heavy chain. Together, these results suggest that EVs enter cells predominantly via clathrin-independent endocytosis and macropinocytosis. Identification of EV components that promote their uptake via pathways that lead to functional cargo transfer might allow development of more efficient therapeutics through EV-inspired engineering.
Collapse
Affiliation(s)
- Helena Costa Verdera
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Jerney J Gitz-Francois
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Raymond M Schiffelers
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Pieter Vader
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| |
Collapse
|
100
|
Iram S, Zahera M, Khan S, Khan I, Syed A, Ansary AA, Ameen F, Shair OHM, Khan MS. Gold nanoconjugates reinforce the potency of conjugated cisplatin and doxorubicin. Colloids Surf B Biointerfaces 2017; 160:254-264. [PMID: 28942160 DOI: 10.1016/j.colsurfb.2017.09.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 12/22/2022]
Abstract
Osteosarcoma or osteogenic sarcoma is the most common and prevalent cancerous tumor of bone and occurs especially in children and teens. Recent treatment strategy includes a combination of both chemotherapy and surgeries. Although, the use of single drug-based chemotherapy treatment remains unsatisfactory. Therefore, combinatorial therapy has emerged as a potential strategy for treatment with limited side- effects. Here, we evaluated the combinatorial anticancerous effect of cisplatin (CIS) and doxorubicin (DOX) bioconjugated bromelain encapsulated gold nanoparticles (B-AuNPs conjugated CIS and DOX) in the treatment of osteosarcoma. The synthesized B-AuNPs conjugated CIS and DOX were characterized by various characterization techniques like UV-vis spectroscopy, TEM, DLS and zeta potential to ensure the synthesis, size, shape, size distribution and stability. Drug loading efficiency bioconjugation of CIS and DOX was ensured by UV-vis spectroscopy. Bioconjugation of CIS and DOX was further confirmed using UV-vis spectroscopy, TEM, DLS, Zeta potential and FT-IR analysis. The combinatorial effect of CIS and DOX in B-AuNPs conjugated CIS and DOX showed highly improved potency against MG-63 and Saos-2 cells at a very low concentration where primary osteoblasts didn't show any cytotoxic effect. The apoptotic effect of B-AuNPs conjugated CIS and DOX on osteosarcoma and primary osteoblasts cells were analyzed by increased permeability of the cell membrane, condensed chromatin and deep blue fluorescent condensed nucleus. The results clearly showed that B-AuNPs conjugated CIS and DOX significantly improved the potency of both the chemotherapeutic drugs by delivering them specifically into the nucleus of cancer cells through caveolae-dependent endocytosis. Thus, the greater inhibitory effect of combinatorial drugs (B-AuNPs conjugated CIS and DOX) over single drug based chemotherapy would be of great advantage during osteosarcoma treatment.
Collapse
Affiliation(s)
- Sana Iram
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Manaal Zahera
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Salman Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Imran Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abu Ayoobul Ansary
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, 411008, India
| | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Omar H M Shair
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohd Sajid Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India.
| |
Collapse
|