1
|
Chen C, Wang B, Zhao X, Luo Y, Fu L, Qi X, Ying Z, Chen L, Wang Q, Sun S, Chen D, Kang P. Lithium Promotes Osteogenesis via Rab11a-Facilitated Exosomal Wnt10a Secretion and β-Catenin Signaling Activation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30793-30809. [PMID: 38833412 DOI: 10.1021/acsami.4c04199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Both bone mesenchymal stem cells (BMSCs) and their exosomes suggest promising therapeutic tools for bone regeneration. Lithium has been reported to regulate BMSC function and engineer exosomes to improve bone regeneration in patients with glucocorticoid-induced osteonecrosis of the femoral head. However, the mechanisms by which lithium promotes osteogenesis have not been elucidated. Here, we demonstrated that lithium promotes the osteogenesis of BMSCs via lithium-induced increases in the secretion of exosomal Wnt10a to activate Wnt/β-catenin signaling, whose secretion is correlated with enhanced MARK2 activation to increase the trafficking of the Rab11a and Rab11FIP1 complexes together with exosomal Wnt10a to the plasma membrane. Then, we compared the proosteogenic effects of exosomes derived from lithium-treated or untreated BMSCs (Li-Exo or Con-Exo) both in vitro and in vivo. We found that, compared with Con-Exo, Li-Exo had superior abilities to promote the uptake and osteogenic differentiation of BMSCs. To optimize the in vivo application of these hydrogels, we fabricated Li-Exo-functionalized gelatin methacrylate (GelMA) hydrogels, which are more effective at promoting osteogenesis and bone repair than Con-Exo. Collectively, these findings demonstrate the mechanism by which lithium promotes osteogenesis and the great promise of lithium for engineering BMSCs and their exosomes for bone regeneration, warranting further exploration in clinical practice.
Collapse
Affiliation(s)
- Changjun Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Baoning Wang
- Department of Microbiology, West China of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xin Zhao
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, China
| | - Yue Luo
- Department of Orthopedic Surgery, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Li Fu
- Research Core Facility, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Qi
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhendong Ying
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Liyile Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuru Wang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuo Sun
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dailing Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Pengde Kang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
Wang X, Wang WX. Tracking the Cellular Degradation of Silver Nanoparticles: Development of a Generic Kinetic Model. ACS NANO 2024; 18:13308-13321. [PMID: 38716827 DOI: 10.1021/acsnano.4c03032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Understanding the degradation of nanoparticles (NPs) after crossing the cell plasma membrane is crucial in drug delivery designs and cytotoxicity assessment. However, the key factors controlling the degradable kinetics remain unclear due to the absence of a quantification model. In this study, subcellular imaging of silver nanoparticles (AgNPs) was used to determine the intracellular transfer of AgNPs, and single particle ICP-MS was utilized to track the degradation process. A cellular kinetic model was subsequently developed to describe the uptake, transfer, and degradation behaviors of AgNPs. Our model demonstrated that the intracellular degradation efficiency of AgNPs was much higher than that determined by mimicking testing, and the degradation of NPs was highly influenced by cellular factors. Specifically, deficiencies in Ca or Zn primarily decreased the kinetic dissolution of NPs, while a Ca deficiency also resulted in the retardation of NP transfer. The biological significance of these kinetic parameters was strongly revealed. Our model indicated that the majority of internalized AgNPs dissolved, with the resulting ions being rapidly depurated. The release of Ag ions was largely dependent on the microvesicle-mediated route. By changing the coating and size of AgNPs, the model results suggested that size influenced the transfer of NPs into the degradation process, whereas coating affected the degradation kinetics. Overall, our developed model provides a valuable tool for understanding and predicting the impacts of the physicochemical properties of NPs and the ambient environment on nanotoxicity and therapeutic efficacy.
Collapse
Affiliation(s)
- Xiangrui Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
3
|
Andrian T, Muela Y, Delgado L, Albertazzi L, Pujals S. A super-resolution and transmission electron microscopy correlative approach to study intracellular trafficking of nanoparticles. NANOSCALE 2023; 15:14615-14627. [PMID: 37614108 DOI: 10.1039/d3nr02838k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Nanoparticles (NPs) are used to encapsulate therapeutic cargos and deliver them specifically to the target site. The intracellular trafficking of NPs dictates the NP-cargo distribution within different cellular compartments, and thus governs their efficacy and safety. Knowledge in this field is crucial to understand their biological fate and improve their rational design. However, there is a lack of methods that allow precise localization and quantification of individual NPs within distinct cellular compartments simultaneously. Here, we address this issue by proposing a correlative light and electron microscopy (CLEM) method combining direct stochastic optical reconstruction microscopy (dSTORM) and transmission electron microscopy (TEM). We aim at combining the advantages of both techniques to precisely address NP localization in the context of the cell ultrastructure. Individual fluorescently-labelled poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) NPs were directly visualized by dSTORM and assigned to cellular compartments by TEM. We first tracked NPs along the endo-lysosomal pathway at different time points, then demonstrated the effect of chloroquine on their intracellular distribution (i.e. endosomal escape). The proposed protocol can be applied to fluorescently labelled NPs and/or cargo, including those not detectable by TEM alone. Our studies are of great relevance to obtain important information on NP trafficking, and crucial for the design of more complex nanomaterials aimed at cytoplasmic/nucleic drug delivery.
Collapse
Affiliation(s)
- Teodora Andrian
- Institute for Bioengineering of Catalonia (IBEC), Carrer Baldiri Reixac 15-21, 08024 Barcelona, Spain.
| | - Yolanda Muela
- Electron Cryomicroscopy Unit, Centres Científics i Tecnològics de la Universitat de Barcelona (CCiTUB), Carrer Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Lidia Delgado
- Electron Cryomicroscopy Unit, Centres Científics i Tecnològics de la Universitat de Barcelona (CCiTUB), Carrer Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), Carrer Baldiri Reixac 15-21, 08024 Barcelona, Spain.
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Silvia Pujals
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Carrer Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
4
|
Corci B, Hooiveld O, Dolga AM, Åberg C. Extending the analogy between intracellular motion in mammalian cells and glassy dynamics. SOFT MATTER 2023; 19:2529-2538. [PMID: 36939775 DOI: 10.1039/d2sm01672a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
How molecules, organelles, and foreign objects move within living cells has been studied in organisms ranging from bacteria to human cells. In mammalian cells, in particular, cellular vesicles move across the cell using motor proteins that carry the vesicle down the cytoskeleton to their destination. We have recently noted several similarities between the motion of such vesicles and that in disordered, "glassy", systems, but the generality of this observation remains unclear. Here we follow the motion of mitochondria, the organelles responsible for cell energy production, in mammalian cells over timescales from 50 ms to 70 s. Qualitative observations show that single mitochondria remain within a spatially limited region for extended periods of time, before moving longer distances relatively quickly. The displacement distribution is roughly Gaussian for shorter distances (≲0.05 μm) but exhibits exponentially decaying tails at longer distances (up to 0.40 μm). This behaviour is well-described by a model developed to describe the motion in glassy systems. These observations are extended to in total 3 different objects (mitochondria, lysosomes and nano-sized beads enclosed in vesicles), 3 different mammalian cell types (HEK 293, HeLa, and HT22), from 2 different organisms (human and mouse). Further evidence that supports glass-like characteristics of the motion is a difference between the time it takes to move a longer distance for the first time and subsequent times, as well as a weak ergodicity breaking of the motion. Overall, we demonstrate the ubiquity of glass-like motion in mammalian cells, providing a different perspective on intracellular motion.
Collapse
Affiliation(s)
- Beatrice Corci
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Oscar Hooiveld
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Amalia M Dolga
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
5
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
6
|
Algharib SA, Dawood A, Zhou K, Chen D, Li C, Meng K, Zhang A, Luo W, Ahmed S, Huang L, Xie S. Preparation of chitosan nanoparticles by ionotropic gelation technique: Effects of formulation parameters and in vitro characterization. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132129] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
7
|
Application of Rapid Fluorescence Lifetime Imaging Microscopy (RapidFLIM) to Examine Dynamics of Nanoparticle Uptake in Live Cells. Cells 2022; 11:cells11040642. [PMID: 35203292 PMCID: PMC8870300 DOI: 10.3390/cells11040642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/28/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
A key challenge in nanomedicine stems from the continued need for a systematic understanding of the delivery of nanoparticles in live cells. Complexities in delivery are often influenced by the biophysical characteristics of nanoparticles, where even subtle changes to nanoparticle designs can alter cellular uptake, transport and activity. Close examination of these processes, especially with imaging, offers important insights that can aid in future nanoparticle design or translation. Rapid fluorescence lifetime imaging microscopy (RapidFLIM) is a potentially valuable technology for examining intracellular mechanisms of nanoparticle delivery by directly correlating visual data with changes in the biological environment. To date, applications for this technology in nanoparticle research have not been explored. A PicoQuant RapidFLIM system was used together with commercial silica nanoparticles to follow particle uptake in glioblastoma cells. Importantly, RapidFLIM imaging showed significantly improved image acquisition speeds over traditional FLIM, which enabled the tracking of nanoparticle uptake into subcellular compartments. We determined mean lifetime changes and used this to delineate significant changes in nanoparticle lifetimes (>0.39 ns), which showed clustering of these tracks proximal to both extracellular and nuclear membrane boundaries. These findings demonstrate the ability of RapidFLIM to track, localize and quantify changes in single nanoparticle fluorescence lifetimes and highlight RapidFLIM as a valuable tool for multiparameter visualization and analysis of nanoparticle molecular dynamics in live cells.
Collapse
|
8
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
9
|
de Boer I, Richards CJ, Åberg C. Simultaneous Exposure of Different Nanoparticles Influences Cell Uptake. Pharmaceutics 2022; 14:136. [PMID: 35057032 PMCID: PMC8779877 DOI: 10.3390/pharmaceutics14010136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 02/01/2023] Open
Abstract
Drug delivery using nano-sized carriers holds tremendous potential for curing a range of diseases. The internalisation of nanoparticles by cells, however, remains poorly understood, restricting the possibility for optimising entrance into target cells, avoiding off-target cells and evading clearance. The majority of nanoparticle cell uptake studies have been performed in the presence of only the particle of interest; here, we instead report measurements of uptake when the cells are exposed to two different types of nanoparticles at the same time. We used carboxylated polystyrene nanoparticles of two different sizes as a model system and exposed them to HeLa cells in the presence of a biomolecular corona. Using flow cytometry, we quantify the uptake at both average and individual cell level. Consistent with previous literature, we show that uptake of the larger particles is impeded in the presence of competing smaller particles and, conversely, that uptake of the smaller particles is promoted by competing larger particles. While the mechanism(s) underlying these observations remain(s) undetermined, we are partly able to restrain the likely possibilities. In the future, these effects could conceivably be used to enhance uptake of nano-sized particles used for drug delivery, by administering two different types of particles at the same time.
Collapse
Affiliation(s)
| | | | - Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (I.d.B.); (C.J.R.)
| |
Collapse
|
10
|
Hatoyama Y, Homma Y, Hiragi S, Fukuda M. Establishment and analysis of conditional Rab1- and Rab5-knockout cells using the auxin-inducible degron system. J Cell Sci 2021; 134:273782. [PMID: 34817057 DOI: 10.1242/jcs.259184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022] Open
Abstract
Two small GTPases, Rab1 and Rab5, are key membrane trafficking regulators that are conserved in all eukaryotes. They have recently been found to be essential for cell survival and/or growth in cultured mammalian cells, thereby precluding the establishment of Rab1-knockout (KO) and Rab5-KO cells, making it extremely difficult to assess the impact of complete Rab1 or Rab5 protein depletion on cellular functions. Here, we generated and analyzed cell lines with conditional KO (CKO) of either Rab1 (Rab1A and Rab1B) or Rab5 (Rab5A, Rab5B and Rab5C) by using the auxin-inducible protein degradation system. Rab1 CKO and Rab5 CKO led to eventual cell death from 18 h and 48 h, respectively, after auxin exposure. After acute Rab1 protein depletion, the Golgi stack and ribbon structures were completely disrupted, and endoplasmic reticulum (ER)-to-Golgi trafficking was severely inhibited. Moreover, we discovered a novel Rab1-depletion phenotype: perinuclear clustering of early endosomes and delayed transferrin recycling. In contrast, acute Rab5 protein depletion resulted in loss of early endosomes and late endosomes, but lysosomes appeared to be normal. We also observed a dramatic reduction in the intracellular signals of endocytic cargos via receptor-mediated or fluid-phase endocytosis in Rab5-depleted cells.
Collapse
Affiliation(s)
- Yuki Hatoyama
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shu Hiragi
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
11
|
Åberg C, Piattelli V, Montizaan D, Salvati A. Sources of variability in nanoparticle uptake by cells. NANOSCALE 2021; 13:17530-17546. [PMID: 34652349 PMCID: PMC8552707 DOI: 10.1039/d1nr04690j] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/28/2021] [Indexed: 06/13/2023]
Abstract
Understanding how nano-sized objects are taken up by cells is important for applications within medicine (nanomedicine), as well as to avoid unforeseen hazard due to nanotechnology (nanosafety). Even within the same cell population, one typically observes a large cell-to-cell variability in nanoparticle uptake, raising the question of the underlying cause(s). Here we investigate cell-to-cell variability in polystyrene nanoparticle uptake by HeLa cells, with generalisations of the results to silica nanoparticles and liposomes, as well as to A549 and primary human umbilical vein endothelial cells. We show that uptake of nanoparticles is correlated with cell size within a cell population, thereby reproducing and generalising previous reports highlighting the role of cell size in nanoparticle uptake. By repeatedly isolating (using fluorescence-activated cell sorting) the cells that take up the most and least nanoparticles, respectively, and performing RNA sequencing on these cells separately, we examine the underlying gene expression that contributes to high and low polystyrene nanoparticle accumulation in HeLa cells. We can thereby show that cell size is not the sole driver of cell-to-cell variability, but that other cellular characteristics also play a role. In contrast to cell size, these characteristics are more specific to the object (nanoparticle or protein) being taken up, but are nevertheless highly heterogeneous, complicating their detailed identification. Overall, our results highlight the complexity underlying the cellular features that determine nanoparticle uptake propensity.
Collapse
Affiliation(s)
- Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Valeria Piattelli
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Daphne Montizaan
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Anna Salvati
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
12
|
Cursi L, Vercellino S, McCafferty MM, Sheridan E, Petseva V, Adumeau L, Dawson KA. Multifunctional superparamagnetic nanoparticles with a fluorescent silica shell for the in vitro study of bio-nano interactions at the subcellular scale. NANOSCALE 2021; 13:16324-16338. [PMID: 34570135 DOI: 10.1039/d1nr04582b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Despite the high level of interest in bio-nano interactions, detailed intracellular mechanisms that govern nanoscale recognition and signalling still need to be unravelled. Magnetic nanoparticles (NPs) are valuable tools for elucidating complex intracellular bio-nano interactions. Using magnetic NPs, it is possible to isolate cell compartments that the particles interact with during intracellular trafficking. Studies at the subcellular scale rely heavily on optical microscopy; therefore, combining the advantages of magnetic recovery with excellent imaging properties to allow intracellular NP tracking is of utmost interest for the nanoscience field. However, it is a challenge to prepare highly magnetic NPs with a suitable fluorescence for the fluorescence imaging techniques typically used for biological studies. Here we present the synthesis of biocompatible multifunctional superparamagnetic multicore NPs with a bright fluorescent silica shell. The incorporation of an organic fluorophore in the silica surrounding the magnetic multicore was optimised to enable the particles to be tracked with the most common imaging techniques. To prevent dye loss resulting from silica dissolution in biological environments, which would reduce the time that the particles could be tracked, we added a thin dense encapsulating silica layer to the NPs which is highly stable in biological media. The synthesised multifunctional nanoparticles were evaluated in cell uptake experiments in which their intracellular location could be clearly identified using fluorescence imaging microscopy, even after 3 days. The magnetic properties of the iron oxide core enabled both efficient recovery of the NPs from the intracellular environment and the extraction of cell compartments involved in their intracellular trafficking. Thus, the NPs reported here provide a promising tool for the study of the processes regulating bio-nano interactions.
Collapse
Affiliation(s)
- Lorenzo Cursi
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Silvia Vercellino
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Mura M McCafferty
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Emily Sheridan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Vanya Petseva
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Laurent Adumeau
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
13
|
Garcia Romeu H, Deville S, Salvati A. Time- and Space-Resolved Flow-Cytometry of Cell Organelles to Quantify Nanoparticle Uptake and Intracellular Trafficking by Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100887. [PMID: 34272923 DOI: 10.1002/smll.202100887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/15/2021] [Indexed: 05/20/2023]
Abstract
The design of targeted nanomedicines requires intracellular space- and time-resolved data of nanoparticle distribution following uptake. Current methods to study intracellular trafficking, such as dynamic colocalization by fluorescence microscopy in live cells, are usually low throughput and require extensive analysis of large datasets to quantify colocalization in several individual cells. Here a method based on flow cytometry to easily detect and characterize the organelles in which nanoparticles are internalized and trafficked over time is proposed. Conventional cell fractionation methods are combined with immunostaining and high-sensitivity organelle flow cytometry to get space-resolved data of nanoparticle intracellular distribution. By extracting the organelles at different times, time-resolved data of nanoparticle intracellular trafficking are obtained. The method is validated by determining how nanoparticle size affects the kinetics of arrival to the lysosomes. The results demonstrate that this method allows high-throughput analysis of nanoparticle uptake and intracellular trafficking by cells, therefore it can be used to determine how nanoparticle design affects their intracellular behavior.
Collapse
Affiliation(s)
- Hector Garcia Romeu
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Sarah Deville
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, Groningen, 9713AV, The Netherlands
- Health Unit, Flemish Institute for Technological Research, Boeretang 200, Mol, 2400, Belgium
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, Groningen, 9713AV, The Netherlands
| |
Collapse
|
14
|
Gravely M, Roxbury D. Multispectral Fingerprinting Resolves Dynamics of Nanomaterial Trafficking in Primary Endothelial Cells. ACS NANO 2021; 15:12388-12404. [PMID: 34180232 DOI: 10.1021/acsnano.1c04500] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Intracellular vesicle trafficking involves a complex series of biological pathways used to sort, recycle, and degrade extracellular components, including engineered nanomaterials (ENMs) which gain cellular entry via active endocytic processes. A recent emphasis on routes of ENM uptake has established key physicochemical properties which direct certain mechanisms, yet relatively few studies have identified their effect on intracellular trafficking processes past entry and initial subcellular localization. Here, we developed and applied an approach where single-walled carbon nanotubes (SWCNTs) play a dual role-that of an ENM undergoing intracellular processing, in addition to functioning as the signal transduction element reporting these events in individual cells with single organelle resolution. We used the exceptional optical properties exhibited by noncovalent hybrids of single-stranded DNA and SWCNTs (DNA-SWCNTs) to report the progression of intracellular processing events via two orthogonal hyperspectral imaging approaches of near-infrared (NIR) fluorescence and resonance Raman scattering. A positive correlation between fluorescence and G-band intensities was uncovered within single cells, while exciton energy transfer and eventual aggregation of DNA-SWCNTs were observed to scale with increasing time after internalization. An analysis pipeline was developed to colocalize and deconvolute the fluorescence and Raman spectra of subcellular regions of interest (ROIs), allowing for single-chirality component spectra to be obtained with submicron spatial resolution. This approach uncovered correlations between DNA-SWCNT concentration, dielectric modulation, and irreversible aggregation within single intracellular vesicles. An immunofluorescence assay was designed to directly observe the DNA-SWCNTs in labeled endosomal vesicles, revealing a distinct relationship between the physical state of organelle-bound DNA-SWCNTs and the dynamic luminal conditions during endosomal maturation processes. Finally, we trained a machine learning algorithm to predict endosome type using the Raman spectra of the vesicle-bound DNA-SWCNTs, enabling major components in the endocytic pathway to be simultaneously visualized using a single intracellular reporter.
Collapse
Affiliation(s)
- Mitchell Gravely
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Daniel Roxbury
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| |
Collapse
|
15
|
Vtyurina N, Åberg C, Salvati A. Imaging of nanoparticle uptake and kinetics of intracellular trafficking in individual cells. NANOSCALE 2021; 13:10436-10446. [PMID: 34076024 PMCID: PMC8211015 DOI: 10.1039/d1nr00901j] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/24/2021] [Indexed: 05/02/2023]
Abstract
Live cell imaging is a powerful tool to understand how nano-sized objects, such as the drug carriers used for nanomedicine applications, are taken up and trafficked by cells. Here we visualized human HeLa cells as they took up and trafficked nanoparticles of different sizes and quantified nanoparticle colocalization with different fluorescently-labelled intracellular compartments over time. This allowed us to obtain kinetic profiles of nanoparticle transport towards the lysosomes in individual cells. With a simple theoretical model, we determined the typical departure time of nanoparticles from the cell membrane and typical lysosome arrival time. We compared these kinetics parameters for nanoparticles of different sizes and determined how they vary in individual cells. We also performed a similar analysis for early endocytic compartments through which nanoparticles transit and discuss challenges in quantifying the colocalization in this case. The results show a high variability in intracellular trafficking kinetics between individual cells. Additionally, they help us to understand how nanoparticle properties affect their cellular uptake and intracellular distribution kinetics.
Collapse
Affiliation(s)
- Natalia Vtyurina
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| | - Christoffer Åberg
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| |
Collapse
|
16
|
Escaping the endosome: assessing cellular trafficking mechanisms of non-viral vehicles. J Control Release 2021; 335:465-480. [PMID: 34077782 DOI: 10.1016/j.jconrel.2021.05.038] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022]
Abstract
Non-viral vehicles hold therapeutic promise in advancing the delivery of a variety of cargos in vitro and in vivo, including small molecule drugs, biologics, and especially nucleic acids. However, their efficacy at the cellular level is limited by several delivery barriers, with endolysosomal degradation being most significant. The entrapment of vehicles and their cargo in the acidified endosome prevents access to the cytosol, nucleus, and other subcellular compartments. Understanding the factors that contribute to uptake and intracellular trafficking, especially endosomal entrapment and release, is key to overcoming delivery obstacles within cells. In this review, we summarize and compare experimental techniques for assessing the extent of endosomal escape of a variety of non-viral vehicles and describe proposed escape mechanisms for different classes of lipid-, polymer-, and peptide-based delivery agents. Based on this evaluation, we present forward-looking strategies utilizing information gained from mechanistic studies to inform the rational design of efficient delivery vehicles.
Collapse
|
17
|
Sheridan E, Vercellino S, Cursi L, Adumeau L, Behan JA, Dawson KA. Understanding intracellular nanoparticle trafficking fates through spatiotemporally resolved magnetic nanoparticle recovery. NANOSCALE ADVANCES 2021; 3:2397-2410. [PMID: 36134166 PMCID: PMC9419038 DOI: 10.1039/d0na01035a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/21/2021] [Indexed: 05/08/2023]
Abstract
The field of nanomedicine has the potential to be a game-changer in global health, with possible applications in prevention, diagnostics, and therapeutics. However, despite extensive research focus and funding, the forecasted explosion of novel nanomedicines is yet to materialize. We believe that clinical translation is ultimately hampered by a lack of understanding of how nanoparticles really interact with biological systems. When placed in a biological environment, nanoparticles adsorb a biomolecular layer that defines their biological identity. The challenge for bionanoscience is therefore to understand the evolution of the interactions of the nanoparticle-biomolecules complex as the nanoparticle is trafficked through the intracellular environment. However, to progress on this route, scientists face major challenges associated with isolation of specific intracellular compartments for analysis, complicated by the diversity of trafficking events happening simultaneously and the lack of synchronization between individual events. In this perspective article, we reflect on how magnetic nanoparticles can help to tackle some of these challenges as part of an overall workflow and act as a useful platform to investigate the bionano interactions within the cell that contribute to this nanoscale decision making. We discuss both established and emerging techniques for the magnetic extraction of nanoparticles and how they can potentially be used as tools to study the intracellular journey of nanomaterials inside the cell, and their potential to probe nanoscale decision-making events. We outline the inherent limitations of these techniques when investigating particular bio-nano interactions along with proposed strategies to improve both specificity and resolution. We conclude by describing how the integration of magnetic nanoparticle recovery with sophisticated analysis at the single-particle level could be applied to resolve key questions for this field in the future.
Collapse
Affiliation(s)
- Emily Sheridan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
| | - Silvia Vercellino
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Biomolecular and Biomedical Science, University College Dublin Belfield Dublin 4 Ireland
| | - Lorenzo Cursi
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
| | - Laurent Adumeau
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
| | - James A Behan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry, University College Dublin Belfield Dublin 4 Ireland
| |
Collapse
|
18
|
Åberg C. Kinetics of nanoparticle uptake into and distribution in human cells. NANOSCALE ADVANCES 2021; 3:2196-2212. [PMID: 36133761 PMCID: PMC9416924 DOI: 10.1039/d0na00716a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/12/2021] [Indexed: 05/17/2023]
Abstract
Whether one wishes to optimise drug delivery using nano-sized carriers or avoid hazard posed by engineered nanomaterials, the kinetics of nanoparticle uptake into human cells and their subsequent intracellular distribution is key. Unique properties of the nanoscale implies that such nanoparticles are taken up and trafficked in a different fashion compared to molecular species. In this review, we discuss in detail how to describe the kinetics of nanoparticle uptake and intracellular distribution, using previous studies for illustration. We also cover the extracellular kinetics, particle degradation, endosomal escape and cell division, ending with an outlook on the future of kinetic studies.
Collapse
Affiliation(s)
- Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen Antonius Deusinglaan 1 9713AV Groningen The Netherlands
| |
Collapse
|
19
|
Åberg C, Poolman B. Glass-like characteristics of intracellular motion in human cells. Biophys J 2021; 120:2355-2366. [PMID: 33887228 DOI: 10.1016/j.bpj.2021.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022] Open
Abstract
The motion in the cytosol of microorganisms such as bacteria and yeast has been observed to undergo a dramatic slowing down upon cell energy depletion. These observations have been interpreted as the motion being "glassy," but whether this notion is useful also for active, motor-protein-driven transport in eukaryotic cells is less clear. Here, we use fluorescence microscopy of beads in human (HeLa) cells to probe the motion of membrane-surrounded structures that are carried along the cytoskeleton by motor proteins. Evaluating several hallmarks of glassy dynamics, we show that at short length scales, the motion is heterogeneous, is nonergodic, is well described by a model for the displacement distribution in glassy systems, and exhibits a decoupling of the exchange and persistence times. Overall, these results suggest that the short length scale behavior of objects that can be transported actively by motor proteins in human cells shares features with the motion in glassy systems.
Collapse
Affiliation(s)
- Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Biochemistry, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands.
| | - Bert Poolman
- Department of Biochemistry, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
20
|
Dawson KA, Yan Y. Current understanding of biological identity at the nanoscale and future prospects. NATURE NANOTECHNOLOGY 2021; 16:229-242. [PMID: 33597736 DOI: 10.1038/s41565-021-00860-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Nanoscale objects are processed by living organisms using highly evolved and sophisticated endogenous cellular networks, specifically designed to manage objects of this size. While these processes potentially allow nanostructures unique access to and control over key biological machineries, they are also highly protected by cell or host defence mechanisms at all levels. A thorough understanding of bionanoscale recognition events, including the molecules involved in the cell recognition machinery, the nature of information transferred during recognition processes and the coupled downstream cellular processing, would allow us to achieve a qualitatively novel form of biological control and advanced therapeutics. Here we discuss evolving fundamental microscopic and mechanistic understanding of biological nanoscale recognition. We consider the interface between a nanostructure and a target cell membrane, outlining the categories of nanostructure properties that are recognized, and the associated nanoscale signal transduction and cellular programming mechanisms that constitute biological recognition.
Collapse
Affiliation(s)
- Kenneth A Dawson
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin, Ireland.
| | - Yan Yan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin, Ireland.
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
21
|
Gräfe C, Müller EK, Gresing L, Weidner A, Radon P, Friedrich RP, Alexiou C, Wiekhorst F, Dutz S, Clement JH. Magnetic hybrid materials interact with biological matrices. PHYSICAL SCIENCES REVIEWS 2020. [DOI: 10.1515/psr-2019-0114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Abstract
Magnetic hybrid materials are a promising group of substances. Their interaction with matrices is challenging with regard to the underlying physical and chemical mechanisms. But thinking matrices as biological membranes or even structured cell layers they become interesting with regard to potential biomedical applications. Therefore, we established in vitro blood-organ barrier models to study the interaction and processing of superparamagnetic iron oxide nanoparticles (SPIONs) with these cellular structures in the presence of a magnetic field gradient. A one-cell-type–based blood-brain barrier model was used to investigate the attachment and uptake mechanisms of differentially charged magnetic hybrid materials. Inhibition of clathrin-dependent endocytosis and F-actin depolymerization led to a dramatic reduction of cellular uptake. Furthermore, the subsequent transportation of SPIONs through the barrier and the ability to detect these particles was of interest. Negatively charged SPIONs could be detected behind the barrier as well as in a reporter cell line. These observations could be confirmed with a two-cell-type–based blood-placenta barrier model. While positively charged SPIONs heavily interact with the apical cell layer, neutrally charged SPIONs showed a retarded interaction behavior. Behind the blood-placenta barrier, negatively charged SPIONs could be clearly detected. Finally, the transfer of the in vitro blood-placenta model in a microfluidic biochip allows the integration of shear stress into the system. Even without particle accumulation in a magnetic field gradient, the negatively charged SPIONs were detectable behind the barrier. In conclusion, in vitro blood-organ barrier models allow the broad investigation of magnetic hybrid materials with regard to biocompatibility, cell interaction, and transfer through cell layers on their way to biomedical application.
Collapse
Affiliation(s)
- Christine Gräfe
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Elena K. Müller
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Lennart Gresing
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Andreas Weidner
- Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau , Ilmenau , Germany
| | - Patricia Radon
- Physikalisch-Technische Bundesanstalt , Berlin , Germany
| | - Ralf P. Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON) , Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen , Erlangen , Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON) , Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen , Erlangen , Germany
| | | | - Silvio Dutz
- Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau , Ilmenau , Germany
| | - Joachim H. Clement
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| |
Collapse
|
22
|
Li W, Suarato G, Cathcart JM, Sargunas PR, Meng Y. Design, characterization, and intracellular trafficking of biofunctionalized chitosan nanomicelles. Biointerphases 2020; 15:061003. [PMID: 33187397 PMCID: PMC7666618 DOI: 10.1116/6.0000380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
The hydrophobically modified glycol chitosan (HGC) nanomicelle has received increasing attention as a promising platform for the delivery of chemotherapeutic drugs. To improve the tumor selectivity of HGC, here an avidin and biotin functionalization strategy was applied. The hydrodynamic diameter of the biotin-avidin-functionalized HGC (cy5.5-HGC-B4F) was observed to be 104.7 nm, and the surface charge was +3.1 mV. Confocal and structured illumination microscopy showed that at 0.1 mg/ml, cy5.5-HGC-B4F nanomicelles were distributed throughout the cytoplasm of MDA-MB-231 breast cancer cells after 2 h of exposure without significant cytotoxicity. To better understand the intracellular fate of the nanomicelles, entrapment studies were performed and demonstrated that some cy5.5-HGC-B4F nanomicelles were capable of escaping endocytic vesicles, likely via the proton sponge effect. Quantitative analysis of the movements of endosomes in living cells revealed that the addition of HGC greatly enhanced the motility of endosomal compartments, and the nanomicelles were transported by early and late endosomes from cell periphery to the perinuclear region. Our results validate the importance of using live-cell imaging to quantitatively assess the dynamics and mechanisms underlying the complex endocytic pathways of nanosized drug carriers.
Collapse
Affiliation(s)
- Weiyi Li
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Giulia Suarato
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Jillian M. Cathcart
- Department of Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York 11794
| | - Paul R. Sargunas
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| | - Yizhi Meng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794
| |
Collapse
|
23
|
Hifni B, Khan M, Devereux SJ, Byrne MH, Quinn SJ, Simpson JC. Investigation of the Cellular Destination of Fluorescently Labeled Carbon Nanohorns in Cultured Cells. ACS APPLIED BIO MATERIALS 2020; 3:6790-6801. [PMID: 35019342 DOI: 10.1021/acsabm.0c00748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The high surface area, facile functionalization, and biocompatibility of carbon nanohorns (CNHs) make them attractive for many applications, including drug delivery. The cellular destination of nanomaterials dictates both the therapeutic application and the potential toxicity. Identifying the uptake mechanism is challenging as several endocytic pathways have been identified that facilitate cellular entry. Here, the cellular uptake of fluorescently labeled CNHs was assessed by utilizing quantitative cell-based assays to determine the factors influencing how internalization occurs and the destinations they reach in HeLa cells. Cell viability assays suggest that about 80% of the cells remained viable even at the highest concentration of 20 μg/mL exposure to CNHs. Uptake studies revealed that when pulse-chase conditions were applied, CNHs were seen to be localized both at the cell periphery and in a juxtanuclear pattern inside HeLa cells, in the latter case colocalizing with the lysosomal marker LAMP1. RNA interference studies, using a panel of RNA tools to individually deplete key molecules associated with the endocytic machinery, failed to block the internalization of CNHs into cells, suggesting that multiple mechanisms of endocytosis are used by this particle type.
Collapse
Affiliation(s)
- Badriah Hifni
- School of Chemistry, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland.,School of Biology & Environmental Science, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| | - Mona Khan
- School of Chemistry, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| | - Stephen J Devereux
- School of Chemistry, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| | - Maria H Byrne
- School of Chemistry, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| | - Susan J Quinn
- School of Chemistry, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| | - Jeremy C Simpson
- School of Biology & Environmental Science, University College Dublin, Belfield, Dublin 4 D04 N2E5, Ireland
| |
Collapse
|
24
|
Zhang RL, Pratiwi FW, Chen BC, Chen P, Wu SH, Mou CY. Simultaneous Single-Particle Tracking and Dynamic pH Sensing Reveal Lysosome-Targetable Mesoporous Silica Nanoparticle Pathways. ACS APPLIED MATERIALS & INTERFACES 2020; 12:42472-42484. [PMID: 32657564 DOI: 10.1021/acsami.0c07917] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Nanoparticle (NP)-based targeted drug delivery is intended to transport therapeutically active molecules to specific cells and particular intracellular compartments. However, there is limited knowledge regarding the complete route of NPs in this targeting scenario. In this study, simultaneously performing motion and dynamic pH sensing using single-particle tracking (SPT) leads to an alternative method of gaining insights into the mesoporous silica nanoparticle's (MSN) journey in targeting lysosome. Two different pH-sensitive dyes and a reference dye are incorporated into mesoporous silica nanoparticles (MSNs) via co-condensation to broaden the measurable pH range (pH 4-7.5) of the nanoprobe. The phosphonate, amine, and lysosomal sorting peptides (YQRLGC) are conjugated onto the MSN's surface to study intracellular nano-biointeractions of two oppositely charged and lysosome-targetable MSNs. The brightness and stability of these MSNs allow their movement and dynamic pH evolution during their journey to be simultaneously monitored in real time. Importantly, a multidimensional analysis of MSN's movement and local pH has revealed new model intracellular dynamic states and distributions of MSNs, previously inaccessible when using single parameters alone. A key result is that YQRLGC-conjugated MSNs took an alternative route to target lysosomes apart from the traditional one, which sped up to 4 h and enhanced their targeting efficiency (up to 32%). The findings enrich our understanding of the intracellular journey of MSNs. This study offers complementary information on correlating the surface design with the full pathway of nanoparticles to achieve targeted delivery of therapeutic payload.
Collapse
Affiliation(s)
- Rong-Lin Zhang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Feby Wijaya Pratiwi
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Si-Han Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, 250 Wu Xinyi Street, Taipei 11031, Taiwan
| | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, 250 Wu Xinyi Street, Taipei 11031, Taiwan
| |
Collapse
|
25
|
Thomsen T, Ayoub AB, Psaltis D, Klok HA. Fluorescence-Based and Fluorescent Label-Free Characterization of Polymer Nanoparticle Decorated T Cells. Biomacromolecules 2020; 22:190-200. [PMID: 32869972 DOI: 10.1021/acs.biomac.0c00969] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cells are attractive carriers for the transport and delivery of nanoparticulate cargo. The use of cell-based carriers allows one to enhance control over the biodistribution of drug-loaded polymers and polymer nanoparticles. One key element in the development of cell-based delivery systems is the loading of the cell-based carrier with the nanoparticle cargo, which can be achieved either by internalization of the payload or by immobilization on the cell surface. The surface modification of cells with nanoparticles or the internalization of nanoparticles by cells is usually monitored with fluorescence-based techniques, such as flow cytometry and confocal microscopy. In spite of the widespread use of these techniques, the use of fluorescent labels also poses some risks and has several drawbacks. Fluorescent dyes may bleach, or leach from, the nanoparticles or alter the physicochemical properties of nanoparticles and their interactions with and uptake by cells. Using poly(d,l-lactic acid) nanoparticles that are loaded with Coumarin 6, BODIPY 493/503, or DiO dyes as a model system, this paper demonstrates that the use of physically entrapped fluorescent labels can lead to false negative or erroneous results. The use of nanoparticles that contain covalently tethered fluorescent dyes instead was found to provide a robust approach to monitor cell surface conjugation reactions and to quantitatively analyze nanoparticle-decorated cells. Finally, it is shown that optical diffraction tomography is an attractive, alternative technique for the characterization of nanoparticle-decorated cells, which obviates the need for fluorescent labels.
Collapse
Affiliation(s)
- Tanja Thomsen
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Ahmed B Ayoub
- Institute of Microengineering, Optics Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment BM, Station 17, CH-1015 Lausanne, Switzerland
| | - Demetri Psaltis
- Institute of Microengineering, Optics Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment BM, Station 17, CH-1015 Lausanne, Switzerland
| | - Harm-Anton Klok
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
26
|
Raj EN, Lin Y, Chen C, Liu K, Chao J. Selective Autophagy Pathway of Nanoparticles and Nanodrugs: Drug Delivery and Pathophysiological Effects. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Emmanuel Naveen Raj
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Yu‐Wei Lin
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Chien‐Hung Chen
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Kuang‐Kai Liu
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Jui‐I Chao
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
- Center For Intelligent Drug Systems and Smart Bio‐devices National Chiao Tung University Hsinchu 30068 Taiwan
| |
Collapse
|
27
|
Borkowska M, Siek M, Kolygina DV, Sobolev YI, Lach S, Kumar S, Cho YK, Kandere-Grzybowska K, Grzybowski BA. Targeted crystallization of mixed-charge nanoparticles in lysosomes induces selective death of cancer cells. NATURE NANOTECHNOLOGY 2020; 15:331-341. [PMID: 32203435 DOI: 10.1038/s41565-020-0643-3] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/14/2020] [Indexed: 05/28/2023]
Abstract
Lysosomes have become an important target for anticancer therapeutics because lysosomal cell death bypasses the classical caspase-dependent apoptosis pathway, enabling the targeting of apoptosis- and drug-resistant cancers. However, only a few small molecules-mostly repurposed drugs-have been tested so far, and these typically exhibit low cancer selectivity, making them suitable only for combination therapies. Here, we show that mixed-charge nanoparticles covered with certain ratios of positively and negatively charged ligands can selectively target lysosomes in cancerous cells while exhibiting only marginal cytotoxicity towards normal cells. This selectivity results from distinct pH-dependent aggregation events, starting from the formation of small, endocytosis-prone clusters at cell surfaces and ending with the formation of large and well-ordered nanoparticle assemblies and crystals inside cancer lysosomes. These assemblies cannot be cleared by exocytosis and cause lysosome swelling, which gradually disrupts the integrity of lysosomal membranes, ultimately impairing lysosomal functions and triggering cell death.
Collapse
Affiliation(s)
- Magdalena Borkowska
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Marta Siek
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Diana V Kolygina
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Yaroslav I Sobolev
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Slawomir Lach
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Sumit Kumar
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Kristiana Kandere-Grzybowska
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea.
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea.
| | - Bartosz A Grzybowski
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Republic of Korea.
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea.
| |
Collapse
|
28
|
Guggenheim EJ, Rappoport JZ, Lynch I. Mechanisms for cellular uptake of nanosized clinical MRI contrast agents. Nanotoxicology 2020; 14:504-532. [PMID: 32037933 DOI: 10.1080/17435390.2019.1698779] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Engineered Nanomaterials (NMs), such as Superparamagnetic Iron Oxide Nanoparticles (SPIONs), offer significant benefits in a wide range of applications, including cancer diagnostic and therapeutic strategies. However, the use of NMs in biomedicine raises safety concerns due to lack of knowledge on possible biological interactions and effects. The initial basis for using SPIONs as biomedical MRI contrast enhancement agents was the idea that they are selectively taken up by macrophage cells, and not by the surrounding cancer cells. To investigate this claim, we analyzed the uptake of SPIONs into well-established cancer cell models and benchmarked this against a common macrophage cell model. In combination with fluorescent labeling of compartments and siRNA silencing of various proteins involved in common endocytic pathways, the mechanisms of internalization of SPIONs in these cell types has been ascertained utilizing reflectance confocal microscopy. Caveolar mediated endocytosis and macropinocytosis are both implicated in SPION uptake into cancer cells, whereas in macrophage cells, a clathrin-dependant route appears to predominate. Colocalization studies confirmed the eventual fate of SPIONs as accumulation in the degradative lysosomes. Dissolution of the SPIONs within the lysosomal environment has also been determined, allowing a fuller understanding of the cellular interactions, uptake, trafficking and effects of SPIONs within a variety of cancer cells and macrophages. Overall, the behavior of SPIONS in non-phagocytotic cell lines is broadly similar to that in the specialist macrophage cells, although some differences in the uptake patterns are apparent.
Collapse
Affiliation(s)
- Emily J Guggenheim
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Joshua Z Rappoport
- Center for Advanced Microscopy, and Nikon Imaging Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Core Technologies for Life Sciences, Boston College, MA, USA
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
29
|
Grupi A, Ashur I, Degani-Katzav N, Yudovich S, Shapira Z, Marzouq A, Morgenstein L, Mandel Y, Weiss S. Interfacing the Cell with "Biomimetic Membrane Proteins". SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903006. [PMID: 31765076 DOI: 10.1002/smll.201903006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/30/2019] [Indexed: 06/10/2023]
Abstract
Integral membrane proteins mediate a myriad of cellular processes and are the target of many therapeutic drugs. Enhancement and extension of the functional scope of membrane proteins can be realized by membrane incorporation of engineered nanoparticles designed for specific diagnostic and therapeutic applications. In contrast to hydrophobic insertion of small amphiphilic molecules, delivery and membrane incorporation of particles on the nanometric scale poses a crucial barrier for technological development. In this perspective, the transformative potential of biomimetic membrane proteins (BMPs), current state of the art, and the barriers that need to be overcome in order to advance the field are discussed.
Collapse
Affiliation(s)
- Asaf Grupi
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Idan Ashur
- Agricultural Research Organization, The Volcani Center, Institute of Agricultural Engineering, Rishon LeZion, 7505101, Israel
| | - Nurit Degani-Katzav
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shimon Yudovich
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Zehavit Shapira
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Adan Marzouq
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Lion Morgenstein
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Yossi Mandel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- School of Optometry and Vision Science, Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shimon Weiss
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
30
|
Thalla M, Kant K, Dalchand, Rawat R, Banerjee S. Merged experimental guided computational strategy toward tuberculosis treatment mediated by alveolar macrophages mannose receptor. J Biomol Struct Dyn 2019; 38:5195-5203. [PMID: 31779532 DOI: 10.1080/07391102.2019.1697369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Macrophage mannose receptor (MMR) is a C-type lectin that regulates the phagocytosis and phagocytosis-lysosome (P-L) fusion in tuberculosis. Mannose-capped lipoarabinomannan, a lipoglycan present at the surface of Mycobacterium tuberculosis, is an important factor in phagocyte attachment and internalization that is specific for MMR. Based on this idea, herein we have designed our experiment to understand the better site-specific delivery against tuberculosis. An experimental outcome was used as a basis to revisit the reverse experimental strategy for tuberculosis management. Stearic mannose was prepared from stearic acid incubation with the D-mannose. Interestingly, stearic mannose explained its internalization via stimulating actin-mediated phagocytic pathway of MMR experimentally. Following, an in silico strategy towards hypothetical designing of various mannose-stearyl conjugates (SBKK1-7) against tuberculosis, as binding promoter of MMR (PDB: 1EGI), was carried out using molecular docking and dynamics approaches. Overall, SPKK-5 viz. ortho stearic mannose showed a higher binding affinity with notable H-bonding and hydrophobic interactions. Pharmacokinetic and toxicity examinations illustrated an ideal range of descriptors values for apex screened compounds. Molecular dynamics simulations have confirmed its significant intactness with the MMR. Ultimately, the whole effort led to the identification of promising hit (SBKK-5), which positively correlates with the experimental work and furthermore need to explore its novel drug delivery systems with improved anti-tubercular therapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maharshi Thalla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Kamal Kant
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Dalchand
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Ravi Rawat
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Subham Banerjee
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| |
Collapse
|
31
|
Yang D, Liu D, Deng H, Zhang J, Qin M, Yuan L, Zou X, Shao B, Li H, Dai W, Zhang H, Wang X, He B, Tang X, Zhang Q. Transferrin Functionization Elevates Transcytosis of Nanogranules across Epithelium by Triggering Polarity-Associated Transport Flow and Positive Cellular Feedback Loop. ACS NANO 2019; 13:5058-5076. [PMID: 31034211 DOI: 10.1021/acsnano.8b07231] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Overcoming the epithelial barriers to enhance drug transport is a focused topic for gastrointestinal, intratracheal, intranasal, vaginal, and intrauterine delivery. Nanomedicines with targeting functionization promote such a process owing to specific ligand-receptor interaction. However, compared to the cell uptake of targeting nanotherapies, currently few studies concentrate on their transcytosis including endocytosis for "in" and exocytosis for "out". In fact, the cellular regulatory mechanism for these pathways as well as the principle of ligand's effect on the transcytosis are almost ignored. Here, we fabricated transferrin (Tf) functionalized nanogranules (Tf-NG) as the nanomedicine model and confirmed the difference in polar distributions of Tf receptors (TfRs) between two epithelium models (bipolarity for Caco-2 and unipolarity for MDCK cells). Compared to the nonspecific reference, Tf-conjugation boosted the endocytosis by different pathways in two cell models and transformed the intracellular route of Tf-NG in both cells differently, affecting exocytosis, recycling, and degradation but not the secretion pathway. Only bipolar cells could establish a complete transport flow from "in" to "out", leading to the enhanced transcytosis of Tf-NG. Importantly, epithelia could make responses to Tf-NG transcytosis. Based on the quantitative proteomics, the intracellular trafficking of Tf-NG altered the protein expression profiles, in which the endocytosis- and transcytosis-related proteins were specifically upregulated. Particularly, only bipolar cells could positively feed back to such trafficking via accelerating the subsequent Tf-NG transcytosis. Here, all the cell transport of Tf-NG was polarity associated. In summary, Tf modification elevated the transcytosis of Tf-NG across the epithelium by triggering the polarity-associated transport flow and positive cell feedback loop. These findings provided an insight into the targeting nanodelivery for efficient transport through epithelial barriers.
Collapse
Affiliation(s)
- Dan Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Dechun Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Hailiang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Jian Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Mengmeng Qin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Lan Yuan
- Centre of Medical and Health Analysis , Peking University , Beijing 100191 , China
| | - Xiajuan Zou
- Centre of Medical and Health Analysis , Peking University , Beijing 100191 , China
| | - Bin Shao
- Department of Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) , Peking University Cancer Hospital and Institute , Beijing 100142 , China
| | - Huiping Li
- Department of Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) , Peking University Cancer Hospital and Institute , Beijing 100142 , China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| | - Xing Tang
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences , Peking University , Beijing 100191 , China
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
- State Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing 100191 , China
| |
Collapse
|
32
|
Sun X, Jiang L, Wang C, Sun S, Mei L, Huang L. Systematic investigation of intracellular trafficking behavior of one-dimensional alumina nanotubes. J Mater Chem B 2019; 7:2043-2053. [PMID: 32254808 DOI: 10.1039/c8tb03349h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanotube materials exhibit high drug loading capacity and controlled drug release properties, providing new opportunities for drug delivery. However, the intracellular trafficking paths of 1-dimensional (1D) nanostructured materials are poorly understood compared to their spherical counterparts, impeding the broad application of 1D materials as drug carriers. Here, we report the intracellular trafficking mechanism of nontoxic and biocompatible nanomaterials called anodic alumina nanotubes (AANTs), a model for 1D materials with a geometry that can be precisely engineered. The results indicated that AANTs enter the cells mainly by caveolin endocytosis and micropinocytosis and that cells use a novel macropinocytosis-late endosomes (LEs)-lysosomes route to transport AANTs. Moreover, liposomes (marked by DsRed-Rab18) are fully involved in the classical pathway of early endosomes (EEs)/LEs developing into lysosomes. The AANTs were delivered to the cells via two pathways: slow endocytosis recycling and GLUT4 exocytosis vesicles. The AANTs also induced intracellular autophagy and then degraded via the endolysosomal pathway. Blocking endolysosomal pathways using autophagy inhibitors prevented the degradation of AANTs through lysosomes. Our results add new insights into the pathways and mechanisms of intracellular trafficking of AANTs, and suggest that intracellular trafficking and lysosomal degradation are highly interdependent and important for efficient drug delivery, and should be evaluated together for drug carrier development.
Collapse
Affiliation(s)
- Xiangyu Sun
- Department of Physics, Tsinghua University, Beijing 10008, China.
| | | | | | | | | | | |
Collapse
|
33
|
Zhao P, Chen B, Li L, Wu H, Li Y, Shaneen B, Zhan X, Gu N. Missing-in-metastasis protein promotes internalization of magnetic nanoparticles via association with clathrin light chain and Rab7. Biochim Biophys Acta Gen Subj 2019; 1863:502-510. [PMID: 30528490 PMCID: PMC8218922 DOI: 10.1016/j.bbagen.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/05/2018] [Accepted: 12/04/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Magnetic nanoparticles (MNPs) have been widely used in biomedical applications. Proper control of the duration of MNPs in circulation promises to improve further their applications, in particularly drug delivery. It is known that the uptake of tissue-associated MNPs is mainly carried out by macrophages. Yet, the molecular mechanism to control MNPs internalization in macrophages remains to be elusive. Missing-in-metastasis (MIM) is a scaffolding protein that is highly expressed in macrophages and regulates receptor-mediated endocytosis. We hypothesize that uptake of MNPs may also involve the function of MIM. METHODS We investigated the effect of MIM expression on the intracellular trafficking of MNPs by transmission electronic microscopy, flow cytometry, o-phenanthroline photometric analysis, Perl's staining, immunofluorescence microscopy and co-immunoprecipitation. To explore the molecular events in MIM-mediated MNPs uptake, we examined the effect of MNPs on the interaction of MIM with clathrin, Rab5 and Rab7. RESULTS Uptake of MNPs was significantly enhanced in cells overexpressing MIM. Upon exposure to MNPs, MIM was associated with clathrin light chain in endocytic vesicles and Rab7, a protein that regulates late endosomes. However, MNPs caused dissociation of MIM with Rab5, an early endosome-associated protein. CONCLUSIONS MIM regulates internalization of MNPs via promoting their trafficking from plasma membrane to late endosomes. GENERAL SIGNIFICANCE Our data unveiled a novel pathway which MNPs internalization and intracellular trafficking in macrophages. This new pathway may allow us to control the uptake of MNPs within cells by targeting MIM, thereby improving their medical applications.
Collapse
Affiliation(s)
- Peng Zhao
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China
| | - Bo Chen
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Materials Science and Devices Institute, Suzhou University of Science and Technology, 1 Kerui Road, Suzhou, Jiangsu 215009, PR China
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hao Wu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 210029, PR China
| | - Yan Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China
| | - Baxter Shaneen
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xi Zhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China.
| |
Collapse
|
34
|
Murschhauser A, Röttgermann PJF, Woschée D, Ober MF, Yan Y, Dawson KA, Rädler JO. A high-throughput microscopy method for single-cell analysis of event-time correlations in nanoparticle-induced cell death. Commun Biol 2019; 2:35. [PMID: 30701200 PMCID: PMC6345847 DOI: 10.1038/s42003-019-0282-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022] Open
Abstract
The temporal context of cell death decisions remains generally hidden in ensemble measurements with endpoint readouts. Here, we describe a method to extract event times from fluorescence time traces of cell death-related markers in automated live-cell imaging on single-cell arrays (LISCA) using epithelial A549 lung and Huh7 liver cancer cells as a model system. In pairwise marker combinations, we assess the chronological sequence and delay times of the events lysosomal membrane permeabilization, mitochondrial outer membrane permeabilization and oxidative burst after exposure to 58 nm amino-functionalized polystyrene nanoparticles (PS-NH2 nanoparticles). From two-dimensional event-time scatter plots we infer a lysosomal signal pathway at a low dose of nanoparticles (25 µg mL-1) for both cell lines, while at a higher dose (100 µg mL-1) a mitochondrial pathway coexists in A549 cells, but not in Huh7. In general, event-time correlations provide detailed insights into heterogeneity and interdependencies in signal transmission pathways.
Collapse
Affiliation(s)
- Alexandra Murschhauser
- Faculty of Physics and Center for NanoSciene (CeNS), Ludwig-Maximilians-Universität, Geschwister-Scholl-Platz 1, Munich, 80539 Germany
| | - Peter J. F. Röttgermann
- Faculty of Physics and Center for NanoSciene (CeNS), Ludwig-Maximilians-Universität, Geschwister-Scholl-Platz 1, Munich, 80539 Germany
| | - Daniel Woschée
- Faculty of Physics and Center for NanoSciene (CeNS), Ludwig-Maximilians-Universität, Geschwister-Scholl-Platz 1, Munich, 80539 Germany
| | - Martina F. Ober
- Faculty of Physics and Center for NanoSciene (CeNS), Ludwig-Maximilians-Universität, Geschwister-Scholl-Platz 1, Munich, 80539 Germany
| | - Yan Yan
- Centre for BioNano Interactions, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kenneth A. Dawson
- Centre for BioNano Interactions, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Joachim O. Rädler
- Faculty of Physics and Center for NanoSciene (CeNS), Ludwig-Maximilians-Universität, Geschwister-Scholl-Platz 1, Munich, 80539 Germany
| |
Collapse
|
35
|
|
36
|
Melamed JR, Ioele SA, Hannum AJ, Ullman VM, Day ES. Polyethylenimine-Spherical Nucleic Acid Nanoparticles against Gli1 Reduce the Chemoresistance and Stemness of Glioblastoma Cells. Mol Pharm 2018; 15:5135-5145. [PMID: 30260647 PMCID: PMC6469355 DOI: 10.1021/acs.molpharmaceut.8b00707] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults, with nearly 100% of patients ultimately succumbing to the disease. Median patient survival is 15 months, and no standard of care currently exists for recurrent cases. Glioma stem cells (GSCs), a rare and highly aggressive subpopulation of cells within these tumors, have recently emerged as drivers of tumor initiation and recurrence, and a growing body of evidence suggests that they must be completely eradicated to prevent relapse. Toward this goal, we have developed polyethylenimine-wrapped spherical nucleic acid nanoparticles (PEI-SNAs) targeting Gli1, a transcription factor within the Hedgehog signaling pathway that is crucial for the maintenance of GSCs. Here, we demonstrate that Gli1 PEI-SNAs bind scavenger receptors on GBM cells to undergo endocytosis in a caveolae/lipid raft/dynamin-dependent manner. They further achieve ∼30% silencing of tumor-promoting Hedgehog pathway genes and downstream target genes that promote the aggressive, chemoresistant phenotype of GBM. This produces a 30% decrease in proliferation that correlates with a robust onset of GBM cell senescence as well as an ∼60% decrease in metabolic activity with or without cotreatment with temozolomide (TMZ), the frontline chemotherapy for GBM. Most importantly, Gli1 PEI-SNAs impair the self-renewal capacity of GBM cells as indicated by a 30-40% reduction in the expression of stemness genes and further impair the formation of stem-like neurospheres. They also substantially improve neurosphere chemosensitivity as demonstrated by a 2-fold increase in the fraction of cells undergoing apoptosis in response to low doses of TMZ. These results underscore the potential for siRNA therapeutics targeting Gli1 to reduce GBM resistance to therapy and warrant further development of PEI-SNAs and Gli1-targeted therapies to alleviate drug resistance and recurrence for GBM patients.
Collapse
Affiliation(s)
- Jilian R. Melamed
- Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Stephen A. Ioele
- Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Ariel J. Hannum
- Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Violet M. Ullman
- Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Emily S. Day
- Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Materials Science & Engineering, University of Delaware, Newark, Delaware 19716, United States
- Helen F. Graham Cancer Center and Research Institute, Newark, Delaware 19713, United States
| |
Collapse
|
37
|
Polli JR, Engler FA, Balthasar JP. Physiologically Based Modeling of the Pharmacokinetics of "Catch-and-Release" Anti-Carcinoembryonic Antigen Monoclonal Antibodies in Colorectal Cancer Xenograft Mouse Models. J Pharm Sci 2018; 108:674-691. [PMID: 30321546 DOI: 10.1016/j.xphs.2018.09.037] [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: 08/20/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 10/28/2022]
Abstract
Engineered monoclonal antibodies (mAbs) with pH-sensitive target release, or "catch-and-release" (CAR) binding, have shown promise in decreasing the extent of target-mediated mAb elimination, increasing mAb exposure, and increasing dose potency. This study developed a mechanistic physiologically based pharmacokinetic (PBPK) model to evaluate the effects of pH-sensitive CAR target binding on the disposition of anti-carcinoembryonic antigen (CEA) mAbs in mouse models of colorectal cancer. The PBPK model was qualified by comparing model-predicted plasma concentration-time data with data observed in tumor-bearing mice following the administration of T84.66, a "standard" anti-CEA mAb that demonstrates strong binding at pH 7.4 and 5.5. Further simulations evaluated the effects CAR pH-dependent binding, with decreasing CEA affinity with decreasing pH, on anti-CEA mAb plasma pharmacokinetics. Simulated data were compared with data observed for a novel CAR mAb, 10H6. The PBPK model provided precise parameter estimates, and excellent data characterization (median prediction error 18.4%) following fitting to T84.66 data. Simulations well predicted 10H6 data (median prediction error 21.4%). Sensitivity analyses demonstrated that key determinants of the disposition of CAR mAbs include the following: antigen binding affinity, the rate constant of mAb-CEA dissociation in acidified endosomes, antigen concentration, and the tumor vasculature reflection coefficient.
Collapse
Affiliation(s)
- Joseph Ryan Polli
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14215
| | - Frank A Engler
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14215
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14215.
| |
Collapse
|
38
|
Chiacchiaretta M, Bramini M, Rocchi A, Armirotti A, Giordano E, Vázquez E, Bandiera T, Ferroni S, Cesca F, Benfenati F. Graphene Oxide Upregulates the Homeostatic Functions of Primary Astrocytes and Modulates Astrocyte-to-Neuron Communication. NANO LETTERS 2018; 18:5827-5838. [PMID: 30088941 DOI: 10.1021/acs.nanolett.8b02487] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene-based materials are the focus of intense research efforts to devise novel theranostic strategies for targeting the central nervous system. In this work, we have investigated the consequences of long-term exposure of primary rat astrocytes to pristine graphene (GR) and graphene oxide (GO) flakes. We demonstrate that GR/GO interfere with a variety of intracellular processes as a result of their internalization through the endolysosomal pathway. Graphene-exposed astrocytes acquire a more differentiated morphological phenotype associated with extensive cytoskeletal rearrangements. Profound functional alterations are induced by GO internalization, including the upregulation of inward-rectifying K+ channels and of Na+-dependent glutamate uptake, which are linked to the astrocyte capacity to control the extracellular homeostasis. Interestingly, GO-pretreated astrocytes promote the functional maturation of cocultured primary neurons by inducing an increase in intrinsic excitability and in the density of GABAergic synapses. The results indicate that graphene nanomaterials profoundly affect astrocyte physiology in vitro with consequences for neuronal network activity. This work supports the view that GO-based materials could be of great interest to address pathologies of the central nervous system associated with astrocyte dysfunctions.
Collapse
Affiliation(s)
| | | | | | | | | | - Ester Vázquez
- Departamento de Química Orgánica , Universidad de Castilla La-Mancha , 13071 Ciudad Real , Spain
| | | | - Stefano Ferroni
- Department of Pharmacy and Biotechnology , University of Bologna , 40126 Bologna , Italy
| | - Fabrizia Cesca
- IRCCS Ospedale Policlinico , San Martino, Genova , Italy
| | | |
Collapse
|
39
|
Willmann W, Dringen R. Monitoring of the Cytoskeleton-Dependent Intracellular Trafficking of Fluorescent Iron Oxide Nanoparticles by Nanoparticle Pulse-Chase Experiments in C6 Glioma Cells. Neurochem Res 2018; 43:2055-2071. [PMID: 30196349 DOI: 10.1007/s11064-018-2627-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/21/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Iron oxide nanoparticles (IONPs) are used for various biomedical and therapeutic approaches. To investigate the uptake and the intracellular trafficking of IONPs in neural cells we have performed nanoparticle pulse-chase experiments to visualize the internalization and the fate of fluorescent IONPs in C6 glioma cells and astrocyte cultures. Already a short exposure to IONPs for 10 min at 4 °C (nanoparticle pulse) allowed binding of substantial amounts of nanoparticles to the cells, while internalization of IONPs into the cell was prevented. The uptake of bound IONPs and the intracellular trafficking was started by increasing the temperature to 37 °C (chase period). While hardly any cellular fluorescence nor any iron staining was detectable directly after the nanoparticle pulse, dotted cellular fluorescence and iron patterns appeared already within a few minutes after start of the chase incubation and became intensified in the perinuclear region during further incubation for up to 90 min. Longer chase incubations resulted in separation of the fluorescent coat from the core of the internalized IONPs. Disruption of actin filaments in C6 cells strongly impaired the internalization of IONPs, whereas destabilization of microtubules traped IONP-containing vesicles to the plasma membrane. In conclusion, nanoparticle pulse-chase experiments allowed to synchronize the cellular uptake of fluorescent IONPs and to identify for C6 cells an actin-dependent early and a microtubule-dependent later process in the intracellular trafficking of fluorescent IONPs.
Collapse
Affiliation(s)
- Wiebke Willmann
- Center for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, 28334, Bremen, Germany
- Center for Environmental Research and Sustainable Technology, Leobener Strasse, 28359, Bremen, Germany
| | - Ralf Dringen
- Center for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, 28334, Bremen, Germany.
- Center for Environmental Research and Sustainable Technology, Leobener Strasse, 28359, Bremen, Germany.
| |
Collapse
|
40
|
Byrne HJ, Bonnier F, Casey A, Maher M, McIntyre J, Efeoglu E, Farhane Z. Advancing Raman microspectroscopy for cellular and subcellular analysis: towards in vitro high-content spectralomic analysis. APPLIED OPTICS 2018; 57:E11-E19. [PMID: 30117916 DOI: 10.1364/ao.57.000e11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/15/2018] [Indexed: 06/08/2023]
Abstract
In the confocal mode, Raman microspectroscopy can profile the biochemical content of biological cells at a subcellular level, and any changes to it by exogenous agents, such as therapeutic drugs or toxicants. As an exploration of the potential of the technique as a high-content, label-free analysis technique, this report reviews work to monitor the spectroscopic signatures associated with the uptake and response pathways of commercial chemotherapeutic agents and polymeric nanoparticles by human lung cells. It is demonstrated that the signatures are reproducible and characteristic of the cellular event, and can be used, for example, to identify the mode of action of the agent as well as the subsequent cell death pathway, and even mechanisms of cellular resistance. Data mining approaches are discussed and a spectralomics approach is proposed.
Collapse
|
41
|
Song S, Cong W, Zhou S, Shi Y, Dai W, Zhang H, Wang X, He B, Zhang Q. Small GTPases: Structure, biological function and its interaction with nanoparticles. Asian J Pharm Sci 2018; 14:30-39. [PMID: 32104436 PMCID: PMC7032109 DOI: 10.1016/j.ajps.2018.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Small GTPase is a kind of GTP-binding protein commonly found in eukaryotic cells. It plays an important role in cytoskeletal reorganization, cell polarity, cell cycle progression, gene expression and many other significant events in cells, such as the interaction with foreign particles. Therefore, it is of great scientific significance to understand the biological properties of small GTPases as well as the GTPase-nano interplay, since more and more nanomedicine are supposed to be used in biomedical field. However, there is no review in this aspect. This review summarizes the small GTPases in terms of the structure, biological function and its interaction with nanoparticles. We briefly introduced the various nanoparticles such as gold/silver nanoparticles, SWCNT, polymeric micelles and other nano delivery systems that interacted with different GTPases. These current nanoparticles exhibited different pharmacological effect modes and various target design concepts in the small GTPases study. This will help to elucidate the conclusion that the therapeutic strategy targeting small GTPases might be a new research direction. It is believed that the in-depth study on the functional mechanism of GTPases can provide insights for the design and study of nanomedicines.
Collapse
Affiliation(s)
- Siyang Song
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.,Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Wenshu Cong
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Shurong Zhou
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Yujie Shi
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Wenbing Dai
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Hua Zhang
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Xueqing Wang
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Bing He
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Qiang Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.,Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| |
Collapse
|
42
|
Banerjee S, Roy S, Nath Bhaumik K, Kshetrapal P, Pillai J. Comparative study of oral lipid nanoparticle formulations (LNFs) for chemical stabilization of antitubercular drugs: physicochemical and cellular evaluation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:540-558. [DOI: 10.1080/21691401.2018.1431648] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Subham Banerjee
- Implants, Devices & Drug Delivery Systems (ID3S) Laboratory, Centre for Biodesign & Diagnostics (CBD), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Subhadeep Roy
- Implants, Devices & Drug Delivery Systems (ID3S) Laboratory, Centre for Biodesign & Diagnostics (CBD), Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Department of Pharmaceutical Sciences, School of Bio-Sciences & Bio-Technology, Baba Saheb Bhimrao Ambedkar University, Lucknow, India
| | - Kaushik Nath Bhaumik
- Implants, Devices & Drug Delivery Systems (ID3S) Laboratory, Centre for Biodesign & Diagnostics (CBD), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | | | - Jonathan Pillai
- Implants, Devices & Drug Delivery Systems (ID3S) Laboratory, Centre for Biodesign & Diagnostics (CBD), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| |
Collapse
|
43
|
Efeoglu E, Maher MA, Casey A, Byrne HJ. Toxicological assessment of nanomaterials: the role of in vitro Raman microspectroscopic analysis. Anal Bioanal Chem 2017; 410:1631-1646. [DOI: 10.1007/s00216-017-0812-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/22/2017] [Accepted: 12/06/2017] [Indexed: 12/21/2022]
|
44
|
Silvestri A, Di Silvio D, Llarena I, Murray RA, Marelli M, Lay L, Polito L, Moya SE. Influence of surface coating on the intracellular behaviour of gold nanoparticles: a fluorescence correlation spectroscopy study. NANOSCALE 2017; 9:14730-14739. [PMID: 28948261 DOI: 10.1039/c7nr04640e] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
In the biomedical applications of nanoparticles (NPs), the proper choice of surface chemistry is a crucial aspect in their design. The nature of the coating can heavily impact the interaction of NPs with biomolecules, affect the state of aggregation, and ultimately determine their biological fate. As such, protein corona formation and the aggregation behaviour of gold NPs (Au NPs) are studied here. Au NPs are prepared with four distinct surface functionalisations, namely mercaptosuccinic acid (MSA), N-4-thiobutyroil glucosamine, HS-PEG5000 and HS-alkyl-PEG600. Corona formation, aggregation, and the intracellular behaviour of the Au NPs are then investigated by means of Fluorescence Correlation Spectroscopy (FCS) in cell culture media and in live cells. To evaluate the state of aggregation and the formation of a protein corona, the Au NPs are incubated in cell media and the diffusion coefficient is determined via FCS. The in vitro behaviour is compared with the level of aggregation of the NPs in cells. Diffusion times of the NPs are estimated at different positions in the cell after a one hour incubation period. It is found that the majority of MSA and glucose-Au NPs are present inside the cell as slowly diffusing species with diffusion times (τD) greater than 6000 μs (hydrodynamic diameter >250 nm). PEGylated Au NPs adsorb a small amount of protein and manifest low agglomeration both in media and in living cells. In particular, the HS-alkyl-PEG600 coating shows an excellent correlation between lower protein adsorption, 4-fold lower compared to the MSA coated NPs, and limited intracellular aggregation. In the case of single HS-alkyl-PEG600 coated NPs, it is found that typical intracellular τD values range from 500 to 1500 μs, indicating that these particles display reduced aggregation in the intracellular environment.
Collapse
Affiliation(s)
- A Silvestri
- CNR - ISTM, Nanotechnology Lab., Via G. Fantoli 16/15, 20138, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Aoyama M, Yoshioka Y, Arai Y, Hirai H, Ishimoto R, Nagano K, Higashisaka K, Nagai T, Tsutsumi Y. Intracellular trafficking of particles inside endosomal vesicles is regulated by particle size. J Control Release 2017; 260:183-193. [DOI: 10.1016/j.jconrel.2017.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 05/10/2017] [Accepted: 06/11/2017] [Indexed: 02/04/2023]
|
46
|
Åberg C, Kim JA, Salvati A, Dawson KA. Reply to 'The interface of nanoparticles with proliferating mammalian cells'. NATURE NANOTECHNOLOGY 2017; 12:600-603. [PMID: 28681851 DOI: 10.1038/nnano.2017.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Affiliation(s)
- Christoffer Åberg
- Groningen Institute of Biomolecular Sciences and Biotechnology, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Jong Ah Kim
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Anna Salvati
- Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry and Chemical Biology and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
47
|
Castagnola V, Cookman J, de Araújo JM, Polo E, Cai Q, Silveira CP, Krpetić Ž, Yan Y, Boselli L, Dawson KA. Towards a classification strategy for complex nanostructures. NANOSCALE HORIZONS 2017; 2:187-198. [PMID: 32260640 DOI: 10.1039/c6nh00219f] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The range of possible nanostructures is so large and continuously growing, that collating and unifying the knowledge connected to them, including their biological activity, is a major challenge. Here we discuss a concept that is based on the connection of microscopic features of the nanomaterials to their biological impacts. We also consider what would be necessary to identify the features that control their biological interactions, and make them resemble each other in a biological context.
Collapse
Affiliation(s)
- V Castagnola
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Han Y, Li X, Chen H, Hu X, Luo Y, Wang T, Wang Z, Li Q, Fan C, Shi J, Wang L, Zhao Y, Wu C, Chen N. Real-Time Imaging of Endocytosis and Intracellular Trafficking of Semiconducting Polymer Dots. ACS APPLIED MATERIALS & INTERFACES 2017; 9:21200-21208. [PMID: 28586196 DOI: 10.1021/acsami.7b05662] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Semiconducting polymer dots (Pdots) have shown great promise in biomedical applications, including biosensing, drug delivery, and live imaging of cells and biomolecules. Insight into the mechanism and regulation of cellular uptake and intracellular metabolism of Pdots is important for the development of superior Pdots-based theranostic nanoconjugates. Herein, we performed real-time imaging of endocytosis and intracellular trafficking of a type of fluorescent Pdots that showed excellent biocompatibility in various types of cells. The endocytic routes and kinetics of Pdots were differently regulated in distinct cell types. Following endocytosis, Pdots were transported in vesicles along microtubule and destined for lysosomes. Furthermore, our results revealed exosome-mediated extracellular release of Pdots and have tracked the dynamic process at the single particle level. These results provide new insight into the design of more effective and selective imaging probes as well as drug carriers.
Collapse
Affiliation(s)
- Yuping Han
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Xiaoming Li
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China
| | - Haobin Chen
- Department of Biomedical Engineering, Southern University of Science and Technology , Shenzhen, Guangdong 518055, China
| | - Xingjie Hu
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Yao Luo
- College of Life Sciences, Sichuan University , Chengdu 610064, China
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Ting Wang
- College of Life Sciences, Sichuan University , Chengdu 610064, China
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Zejun Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Qian Li
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Chunhai Fan
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China
| | - Jiye Shi
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
- UCB Pharma , 208 Bath Road, Slough SL1 3WE, United Kingdom
| | - Lihua Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Yun Zhao
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology , Shenzhen, Guangdong 518055, China
| | - Nan Chen
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| |
Collapse
|
49
|
Wang T, Wang L, Li X, Hu X, Han Y, Luo Y, Wang Z, Li Q, Aldalbahi A, Wang L, Song S, Fan C, Zhao Y, Wang M, Chen N. Size-Dependent Regulation of Intracellular Trafficking of Polystyrene Nanoparticle-Based Drug-Delivery Systems. ACS APPLIED MATERIALS & INTERFACES 2017; 9:18619-18625. [PMID: 28497682 DOI: 10.1021/acsami.7b05383] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Nanoparticles (NPs) have shown great promise as intracellular imaging probes or nanocarriers and are increasingly being used in biomedical applications. A detailed understanding of how NPs get "in and out" of cells is important for developing new nanomaterials with improved selectivity and less cytotoxicity. Both physical and chemical characteristics have been proven to regulate the cellular uptake of NPs. However, the exocytosis process and its regulation are less explored. Herein, we investigated the size-regulated endocytosis and exocytosis of carboxylated polystyrene (PS) NPs. PS NPs with a smaller size were endocytosed mainly through the clathrin-dependent pathway, whereas PS NPs with a larger size preferred caveolae-mediated endocytosis. Furthermore, our results revealed exocytosis of larger PS NPs and tracked the dynamic process at the single-particle level. These results indicate that particle size is a key factor for the regulation of intracellular trafficking of NPs and provide new insight into the development of more effective cellular nanocarriers.
Collapse
Affiliation(s)
- Ting Wang
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Lu Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Xiaoming Li
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, China
| | - Xingjie Hu
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Yuping Han
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Yao Luo
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Zejun Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Qian Li
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Ali Aldalbahi
- Chemistry Department, King Saud University , Riyadh 11451, Saudi Arabia
| | - Lihua Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Shiping Song
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Chunhai Fan
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| | - Yun Zhao
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Maolin Wang
- College of Life Sciences, Sichuan University , Chengdu 610064, China
| | - Nan Chen
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, China
| |
Collapse
|
50
|
Ali MRK, Wu Y, Ghosh D, Do BH, Chen K, Dawson MR, Fang N, Sulchek TA, El-Sayed MA. Nuclear Membrane-Targeted Gold Nanoparticles Inhibit Cancer Cell Migration and Invasion. ACS NANO 2017; 11:3716-3726. [PMID: 28333438 PMCID: PMC5519406 DOI: 10.1021/acsnano.6b08345] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Most cancer patients die from metastasis. Recent studies have shown that gold nanoparticles (AuNPs) can slow down the migration/invasion speed of cancer cells and suppress metastasis. Since nuclear stiffness of the cell largely decreases cell migration, our hypothesis is that targeting AuNPs to the cell nucleus region could enhance nuclear stiffness, and therefore inhibit cell migration and invasion. Our results showed that upon nuclear targeting of AuNPs, the ovarian cancer cell motilities decrease significantly, compared with nontargeted AuNPs. Furthermore, using atomic force microscopy, we observed an enhanced cell nuclear stiffness. In order to understand the mechanism of cancer cell migration/invasion inhibition, the exact locations of the targeted AuNPs were clearly imaged using a high-resolution three-dimensional imaging microscope, which showed that the AuNPs were trapped at the nuclear membrane. In addition, we observed a greatly increased expression level of lamin A/C protein, which is located in the inner nuclear membrane and functions as a structural component of the nuclear lamina to enhance nuclear stiffness. We propose that the AuNPs that are trapped at the nuclear membrane both (1) add to the mechanical stiffness of the nucleus and (2) stimulate the overexpression of lamin A/C located around the nuclear membrane, thus increasing nuclear stiffness and slowing cancer cell migration and invasion.
Collapse
Affiliation(s)
- Moustafa R. K. Ali
- Laser Dynamics Lab (LDL), School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Yue Wu
- Laser Dynamics Lab (LDL), School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Deepraj Ghosh
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island 02912, United States
| | - Brian H. Do
- School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Kuangcai Chen
- Department of Chemistry, Georgia State University, P.O. Box 3965, Atlanta, Georgia 30302, United States
| | - Michelle R. Dawson
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island 02912, United States
| | - Ning Fang
- Department of Chemistry, Georgia State University, P.O. Box 3965, Atlanta, Georgia 30302, United States
- Corresponding Authors: , ,
| | - Todd A. Sulchek
- School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
- Corresponding Authors: , ,
| | - Mostafa A. El-Sayed
- Laser Dynamics Lab (LDL), School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
- Corresponding Authors: , ,
| |
Collapse
|