51
|
Weber C, Simon J, Mailänder V, Morsbach S, Landfester K. Preservation of the soft protein corona in distinct flow allows identification of weakly bound proteins. Acta Biomater 2018; 76:217-224. [PMID: 29885856 DOI: 10.1016/j.actbio.2018.05.057] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/25/2018] [Accepted: 05/31/2018] [Indexed: 12/22/2022]
Abstract
Nanocarriers that are used for targeted drug delivery come in contact with biological liquids and subsequently proteins will adsorb to the nanocarriers' surface to form the so called 'protein corona'. The protein corona defines the biological identity and determines the biological response towards the nanocarriers in the body. To make nanomedicine safe and reliable it is required to get a better insight into this protein corona and, therefore, the adsorbed proteins have to be characterized. Currently, centrifugation is the common method to isolate the protein corona for further investigations. However, with this method it is only possible to investigate the strongly bound proteins, also referred to as 'hard protein corona'. Therefore, we want to introduce a new separation technique to separate nanoparticles including the soft protein corona containing also loosely bound proteins for further characterization. The used separation technique is the asymmetric flow field-flow fractionation (AF4). We were able to separate the nanoparticles with proteins forming the soft protein corona and were able to show that in our system only the hard protein corona directly influenced the cell uptake behavior. STATEMENT OF SIGNIFICANCE Currently, there is an ongoing debate whether only strongly bound proteins (hard corona) or also loosely bound proteins (soft corona) contribute to the biological identity of nanocarriers, because up to now isolation of the soft corona was not possible. Here, asymmetric flow field-flow fractionation was used to isolate nanoparticles with a preserved soft corona from the biological medium. This enabled the characterization of the soft corona composition and to evaluate its influence on cellular uptake. For our system we found that only the strongly bound proteins (hard corona) determined cell internalization. This method can now be used to evaluate the impact of the soft corona further and to characterize nanomaterials that cannot be separated from blood plasma by other means.
Collapse
|
52
|
Vasti C, Bonnet LV, Galiano MR, Rojas R, Giacomelli CE. Relevance of protein–protein interactions on the biological identity of nanoparticles. Colloids Surf B Biointerfaces 2018; 166:330-338. [DOI: 10.1016/j.colsurfb.2018.03.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/08/2018] [Accepted: 03/23/2018] [Indexed: 12/27/2022]
|
53
|
Hsu HJ, Han Y, Cheong M, Král P, Hong S. Dendritic PEG outer shells enhance serum stability of polymeric micelles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1879-1889. [PMID: 29782948 DOI: 10.1016/j.nano.2018.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 02/06/2023]
Abstract
A higher surface density of poly(ethylene glycol) (PEG) on polymeric micelles enhances their stability in serum, leading to improved plasma circulation. To obtain fundamental, mechanistic understanding of the PEG effect associated with polymeric architecture/configuration, we have synthesized PEGylated dendron-based copolymers (PDCs) and linear block copolymers (LBCs) with similar molecular weights. These copolymers formed dendron (hyperbranched) and linear micelles, respectively, which were compared in terms of their stabilities in serum, micelle-serum protein interactions, and in vivo biodistributions. Overall, the dendron micelles exhibited a better serum stability (longer half-life) and thus a slower release profile than the linear micelles. Fluorescence quenching assays and molecular dynamics (MD) simulations revealed that the high serum stability of the dendron micelles can be attributed to reduced micelle-serum protein interactions, owing to their dendritic, dense PEG outer shell. These results provide an important design cue for various polymeric micelles and nanoparticles.
Collapse
Affiliation(s)
- Hao-Jui Hsu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI; Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL
| | - Yanxiao Han
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL
| | - Michael Cheong
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL; Department of Physics, University of Illinois at Chicago, Chicago, IL.
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI; Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL; Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul.
| |
Collapse
|
54
|
Ou W, Thapa RK, Jiang L, Soe ZC, Gautam M, Chang JH, Jeong JH, Ku SK, Choi HG, Yong CS, Kim JO. Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy. J Control Release 2018; 281:84-96. [PMID: 29777794 DOI: 10.1016/j.jconrel.2018.05.018] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/28/2018] [Accepted: 05/16/2018] [Indexed: 12/16/2022]
Abstract
Immunosuppression in tumor microenvironments induced by regulatory T (Treg) cells is regarded a critical mechanism of tumor immune escape and poses a major impediment to cancer immunotherapy. In this study, we developed tLyp1 peptide-conjugated hybrid nanoparticles for targeting Treg cells in the tumor microenvironment. The tLyp1 peptide-modified hybrid nanoparticles presented good stability and effective targeting to Treg cells, and they enhanced the effect of imatinib in downregulating Treg cell suppression through inhibition of STAT3 and STAT5 phosphorylation. In addition, an in vivo study revealed high tumor accumulation of the hybrid nanoparticle. Specifically, prolonged survival rate, enhanced tumor inhibition, reduced intratumoral Treg cells, and elevated intratumoral CD8+ T cells against tumor were observed when combined with checkpoint-blockade by using anti-cytotoxic T-lymphocyte antigen-4 antibody. This study provided groundwork for a repertoire of nanoparticle-based drugs for targeting and modulating Treg cell function in the tumor microenvironment and for improving antitumor immunotherapy.
Collapse
Affiliation(s)
- Wenquan Ou
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Raj Kumar Thapa
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Liyuan Jiang
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Zar Chi Soe
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, South Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, South Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea.
| |
Collapse
|
55
|
Hu J, Youssefian S, Obayemi J, Malatesta K, Rahbar N, Soboyejo W. Investigation of adhesive interactions in the specific targeting of Triptorelin-conjugated PEG-coated magnetite nanoparticles to breast cancer cells. Acta Biomater 2018; 71:363-378. [PMID: 29458110 DOI: 10.1016/j.actbio.2018.02.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022]
Abstract
The understanding of adhesive interaction at the nanoscale between functionalized nanoparticles and biological cells is of great importance to develop effective theranostic nanocarriers for targeted cancer therapy. Here, we report a combination of experimental and computational approaches to evaluate the adhesion between Triptorelin (a Luteinizing Hormone-Releasing Hormone (LHRH) agonist)-conjugated poly-(ethylene glycol) (PEG)-coated magnetite nanoparticles (Triptorelin-MNPs) and breast cells. The adhesion forces between Triptorelin-MNPs and normal/cancerous breast cells are obtained using atomic force microscopy. The corresponding work of adhesion is then estimated using Johnson-Kendall-Roberts model. Our results demonstrate that Triptorelin-MNPs have a fourteen-fold greater work of adhesion to breast cancer cells than to normal breast cells. In addition, the work of adhesion between Triptorelin-MNPs and breast cancer cells is found to be three times more than that between unmodified MNPs and breast cancer cells. Hence, the experimental observation indicates that Triptorelin ligands facilitate the specific targeting of breast cancer cells. Furthermore, molecular dynamics simulations are performed to investigate the molecular origins of the adhesive interactions. The simulations reveal that the interactions between molecules (e.g. Triptorelin and PEG) and LHRH receptors are dominated by van der Waals energies, while the interactions of these molecules with cell membrane are dominated by electrostatic interactions. Moreover, both experimental and computational results reveal that PEG serves as an effective coating that enhances adhesive interactions to breast cancer cells that over-express LHRH receptors, while reduces the adhesion to normal breast cells. Our results highlight the potential to develop Triptorelin-MNPs into tumor-specific MRI contrast agents and drug carriers. STATEMENT OF SIGNIFICANCE Systematic investigation of adhesive interactions between functionalized nanoparticles and cancer cells is of great importance in developing effective theranostic nanocarriers for targeted cancer therapy. Herein, we use a combination of atomic force microscopy technique and molecular dynamics simulations approach to explore the adhesive interactions at the nanoscale between Triptorelin-conjugated polyethylene glycol (PEG)-coated magnetite nanoparticles and normal/cancerous breast cells. This study characterizes and quantifies the work of adhesion, as well as adhesion forces, at the nanocarrier/cell interfaces, unravels the molecular origins of adhesive interactions and highlights the effectiveness of PEG coatings and Triptorelin ligands in the specific targeting of breast cancer cells. Our findings expand the fundamental understanding of nanoparticle/cell adhesion and provide guidelines for the design of more rational nanocarriers.
Collapse
|
56
|
Xiao W, Xiong J, Zhang S, Xiong Y, Zhang H, Gao H. Influence of ligands property and particle size of gold nanoparticles on the protein adsorption and corresponding targeting ability. Int J Pharm 2018; 538:105-111. [DOI: 10.1016/j.ijpharm.2018.01.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/20/2017] [Accepted: 01/03/2018] [Indexed: 11/29/2022]
|
57
|
Findlay MR, Freitas DN, Mobed-Miremadi M, Wheeler KE. Machine learning provides predictive analysis into silver nanoparticle protein corona formation from physicochemical properties. ENVIRONMENTAL SCIENCE. NANO 2018; 5:64-71. [PMID: 29881624 PMCID: PMC5986185 DOI: 10.1039/c7en00466d] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Proteins encountered in biological and environmental systems bind to engineered nanomaterials (ENMs) to form a protein corona (PC) that alters the surface chemistry, reactivity, and fate of the ENMs. Complexities such as the diversity of the PC and variation with ENM properties and reaction conditions make the PC population difficult to predict. Here, we support the development of predictive models for PC populations by relating biophysicochemical characteristics of proteins, ENMs, and solution conditions to PC formation using random forest classification. The resulting model offers a predictive analysis into the population of PC proteins in Ag ENM systems of various ENM size and surface coatings. With an area under the receiver operating characteristic curve of 0.83 and F1-score of 0.81, a model with strong performance has been constructed based upon experimental data. The weighted contribution of each variable provides recommendations for mechanistic models based upon protein enrichment classification results. Protein biophysical properties such as pI and weight are weighted heavily. Yet, ENM size, surface charge, and solution ionic strength also proved essential to an accurate model. The model can be readily modified and applied to other ENM PC populations. The model presented here represents the first step toward robust predictions of PC fingerprints.
Collapse
Affiliation(s)
- Matthew R Findlay
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, United States
| | - Daniel N Freitas
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, United States
| | - Maryam Mobed-Miremadi
- Department of Bioengineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, United States
| | - Korin E Wheeler
- Department of Chemistry & Biochemistry, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, United States
| |
Collapse
|
58
|
Naidu PSR, Norret M, Smith NM, Dunlop SA, Taylor NL, Fitzgerald M, Iyer KS. The Protein Corona of PEGylated PGMA-Based Nanoparticles is Preferentially Enriched with Specific Serum Proteins of Varied Biological Function. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:12926-12933. [PMID: 29022719 DOI: 10.1021/acs.langmuir.7b02568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The composition of the protein corona formed on poly(ethylene glycol)-functionalized (PEGylated) poly(glycidyl methacrylate) (PGMA) nanoparticles (NPs) was qualitatively and quantitatively compared to the protein corona on non-PEGylated PGMA NPs. Despite the reputation of PEGylated NPs for stealth functionality, we demonstrate the preferential enrichment of specific serum proteins of varied biological function in the protein corona on PEGylated NPs when compared to non-PEGylated NPs. Additionally, we suggest that the base material of polymeric NPs plays a role in the preferential enrichment of select serum proteins to the hard corona.
Collapse
Affiliation(s)
| | | | | | | | | | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University and the Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
| | | |
Collapse
|
59
|
|
60
|
Pisani C, Rascol E, Dorandeu C, Gaillard JC, Charnay C, Guari Y, Chopineau J, Armengaud J, Devoisselle JM, Prat O. The species origin of the serum in the culture medium influences the in vitro toxicity of silica nanoparticles to HepG2 cells. PLoS One 2017; 12:e0182906. [PMID: 28796831 PMCID: PMC5552166 DOI: 10.1371/journal.pone.0182906] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/26/2017] [Indexed: 01/08/2023] Open
Abstract
The formation of a protein corona around nanoparticles can influence their toxicity, triggering cellular responses that may be totally different from those elicited by pristine nanoparticles. The main objective of this study was to investigate whether the species origin of the serum proteins forming the corona influences the in vitro toxicity assessment of silica nanoparticles. Coronas were preformed around nanoparticles before cell exposures by incubation in fetal bovine (FBS) or human (HS) serum. The compositions of these protein coronas were assessed by nano-LC MS/MS. The effects of these protein-coated nanoparticles on HepG2 cells were monitored using real-time cell impedance technology. The nanoparticle coronas formed in human or fetal bovine serum comprised many homologous proteins. Using human compared with fetal bovine serum, nanoparticle toxicity in HepG2 cells decreased by 4-fold and 1.5-fold, when used at 50 and 10μg/mL, respectively. It is likely that "markers of self" are present in the serum and are recognized by human cell receptors. Preforming a corona with human serum seems to be more appropriate for in vitro toxicity testing of potential nanocarriers using human cells. In vitro cytotoxicity assays must reflect in vivo conditions as closely as possible to provide solid and useful results.
Collapse
Affiliation(s)
- Cédric Pisani
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
- CEA, Direction de la Recherche Fondamentale—BIAM, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Estelle Rascol
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Christophe Dorandeu
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Jean-Charles Gaillard
- CEA, Direction de la Recherche Fondamentale—IBITECS, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Clarence Charnay
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, Montpellier, France
| | - Yannick Guari
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, Montpellier, France
| | - Joël Chopineau
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
- Université de Nîmes Rue Georges Salan, Nîmes, France
| | - Jean Armengaud
- CEA, Direction de la Recherche Fondamentale—IBITECS, Site de Marcoule, Bagnols-sur-Cèze, France
| | - Jean-Marie Devoisselle
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, Place Eugène Bataillon, Montpellier, France
| | - Odette Prat
- CEA, Direction de la Recherche Fondamentale—BIAM, Site de Marcoule, Bagnols-sur-Cèze, France
| |
Collapse
|
61
|
Engineered polymeric nanoparticles to guide the cellular internalization and trafficking of small interfering ribonucleic acids. J Control Release 2017; 259:3-15. [DOI: 10.1016/j.jconrel.2017.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/29/2022]
|
62
|
Boyes WK, Thornton BLM, Al-Abed SR, Andersen CP, Bouchard DC, Burgess RM, Hubal EAC, Ho KT, Hughes MF, Kitchin K, Reichman JR, Rogers KR, Ross JA, Rygiewicz PT, Scheckel KG, Thai SF, Zepp RG, Zucker RM. A comprehensive framework for evaluating the environmental health and safety implications of engineered nanomaterials. Crit Rev Toxicol 2017; 47:767-810. [DOI: 10.1080/10408444.2017.1328400] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- William K. Boyes
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Brittany Lila M. Thornton
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Souhail R. Al-Abed
- National Risk Management Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Cincinnati, OH, USA
| | - Christian P. Andersen
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Corvallis, OR, USA
| | - Dermont C. Bouchard
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Athens, GA, USA
| | - Robert M. Burgess
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Narragansett, RI, USA
| | - Elaine A. Cohen Hubal
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Kay T. Ho
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Narragansett, RI, USA
| | - Michael F. Hughes
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Kirk Kitchin
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jay R. Reichman
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Corvallis, OR, USA
| | - Kim R. Rogers
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jeffrey A. Ross
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Paul T. Rygiewicz
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Corvallis, OR, USA
| | - Kirk G. Scheckel
- National Risk Management Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Cincinnati, OH, USA
| | - Sheau-Fung Thai
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Richard G. Zepp
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Athens, GA, USA
| | - Robert M. Zucker
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
63
|
Sobczynski DJ, Eniola‐Adefeso O. IgA and IgM protein primarily drive plasma corona-induced adhesion reduction of PLGA nanoparticles in human blood flow. Bioeng Transl Med 2017; 2:180-190. [PMID: 28932819 PMCID: PMC5579729 DOI: 10.1002/btm2.10064] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
The high abundance of immunoglobulins (Igs) in the plasma protein corona on poly(lactic-co-glycolic) acid (PLGA)-based vascular-targeted carriers (VTCs) has previously been shown to reduce their adhesion to activated endothelial cells (aECs) in human blood flow. However, the relative role of individual Ig classes (e.g., IgG, IgA, and IgM) in causing adhesion reduction remains largely unknown. Here, we characterized the influence of specific Ig classes in prescribing the binding efficiency of PLGA nano-sized VTCs in blood flow. Specifically, we evaluated the flow adhesion to aECs of PLGA VTCs with systematic depletion of various Igs in their corona. Adhesion reduction was largely eliminated for PLGA VTCs when all Igs were removed from the corona. Furthermore, re-addition of IgA or IgM to the Igs-depleted corona reinstated the low adhesion of PLGA VTCs, as evidenced by ∼40-70% reduction relative to particles with an Igs-deficient corona. However, re-addition of a high concentration of IgG to the Igs-depleted corona did not cause significant adhesion reduction. Overall, the presented results reveal that PLGA VTC adhesion reduction in blood flows is primarily driven by high adsorption of IgA and IgM in the particle corona. Pre-coating of albumin on PLGA VTCs mitigated the extent of adhesion reduction in plasma for some donors but was largely ineffective in general. Overall, this work may shed light into effective control of protein corona composition, thereby enhancing VTC functionality in vivo for eventual clinical use.
Collapse
Affiliation(s)
| | - Omolola Eniola‐Adefeso
- Dept. of Chemical EngineeringUniversity of MichiganAnn ArborMI48109
- Dept. of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109
- Dept. of Macromolecular Science and EngineeringUniversity of MichiganAnn ArborMI48109
| |
Collapse
|
64
|
Pearson RM, Casey LM, Hughes KR, Miller SD, Shea LD. In vivo reprogramming of immune cells: Technologies for induction of antigen-specific tolerance. Adv Drug Deliv Rev 2017; 114:240-255. [PMID: 28414079 PMCID: PMC5582017 DOI: 10.1016/j.addr.2017.04.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/01/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023]
Abstract
Technologies that induce antigen-specific immune tolerance by mimicking naturally occurring mechanisms have the potential to revolutionize the treatment of many immune-mediated pathologies such as autoimmunity, allograft rejection, and allergy. The immune system intrinsically has central and peripheral tolerance pathways for eliminating or modulating antigen-specific responses, which are being exploited through emerging technologies. Antigen-specific tolerogenic responses have been achieved through the functional reprogramming of antigen-presenting cells or lymphocytes. Alternatively, immune privileged sites have been mimicked using biomaterial scaffolds to locally suppress immune responses and promote long-term allograft survival. This review describes natural mechanisms of peripheral tolerance induction and the various technologies being developed to achieve antigen-specific immune tolerance in vivo. As currently approved therapies are non-specific and carry significant associated risks, these therapies offer significant progress towards replacing systemic immune suppression with antigen-specific therapies to curb aberrant immune responses.
Collapse
Affiliation(s)
- Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Liam M Casey
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 E. Chicago Avenue, Chicago, IL 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA; Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA.
| |
Collapse
|
65
|
Gao J, Lin L, Wei A, Sepúlveda MS. Protein Corona Analysis of Silver Nanoparticles Exposed to Fish Plasma. ENVIRONMENTAL SCIENCE & TECHNOLOGY LETTERS 2017; 4:174-179. [PMID: 31531386 PMCID: PMC6748332 DOI: 10.1021/acs.estlett.7b00074] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanoparticles (NPs) in contact with biological fluids experience changes in surface chemistry that can impact their biodistribution and downstream physiological impact. One such change involves the formation of a protein corona (PC) on the surface of NPs. Here we present a foundational study on PC formation following the incubation of polyvinylpyrrolidone-coated AgNPs (PVP-AgNPs, 50 nm) in the plasma of smallmouth bass (Micropterus dolomieu). PC formation increases with exposure time and is also affected by gender, with AgNPs incubated in male plasma having slightly thinner PCs and less negative zeta potentials than those incubated in female plasma. Proteomic analysis also revealed gender-specific differences in PC composition: in particular, egg-specific proteins (vitellogenin (VTG) and zona pellucida (ZP) were identified only in PCs derived from female plasma, raising the possibility of their roles in AgNP-related reproductive toxicity by promoting their accumulation in developing oocytes.
Collapse
Affiliation(s)
- Jiejun Gao
- Department of Forestry and Natural Resources and Bindley Biosciences Center, Purdue University, West
Lafayette, Indiana, USA
| | - Lu Lin
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Alexander Wei
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
- Corresponding authors: Maria S. Sepúlveda (),
Alexander Wei ()
| | - Maria S. Sepúlveda
- Department of Forestry and Natural Resources and Bindley Biosciences Center, Purdue University, West
Lafayette, Indiana, USA
- Corresponding authors: Maria S. Sepúlveda (),
Alexander Wei ()
| |
Collapse
|
66
|
Monteiro LOF, Fernandes RS, Castro LC, Cardoso VN, Oliveira MC, Townsend DM, Ferretti A, Rubello D, Leite EA, de Barros ALB. Technetium-99m radiolabeled paclitaxel as an imaging probe for breast cancer in vivo. Biomed Pharmacother 2017; 89:146-151. [PMID: 28222395 PMCID: PMC5553547 DOI: 10.1016/j.biopha.2017.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 12/19/2022] Open
Abstract
The high incidence and mortality of breast cancer supports efforts to develop innovative imaging probes to effectively diagnose, evaluate the extent of the tumor, and predict the efficacy of tumor treatments while concurrently and selectively delivering anticancer agents to the cancer tissue. In the present study we described the preparation of technetium-99m (99mTc)-labeled paclitaxel (PTX) and evaluated its feasibility as a radiotracer for breast tumors (4T1) in BALB/c mice. Thin Layer Chromatography (TLC) was used to determine the radiochemical purity and in vitro stability of 99mTc-PTX. PTX micelles showed a unimodal distribution with mean diameter of 13.46±0.06nm. High radiochemical purity (95.8±0.3%) and in vitro stability (over than 95%), up to 24h, were observed. Blood circulation time of 99mTc-PTX was determined in healthy BALB/c mice. 99mTc-PTX decays in a one-phase manner with a half-life of 464.3 minutes. Scintigraphic images and biodistribution were evaluated at 4, 8 and 24h after administration of 99mTc-PTX in 4T1 tumor-bearing mice. The data showed a significant uptake in the liver, spleen and kidneys, due to the importance of these routes for excretion. Moreover, high tumor uptake was achieved, indicated by high tumor-to-muscle ratios. These findings indicate the usefulness of 99mTc-PTX as a radiotracer to identify 4T1 tumor in animal models. In addition, 99mTc-PTX might be used to follow-up treatment protocols in research, being able to provide information about tumor progression after therapy.
Collapse
Affiliation(s)
- Liziane O F Monteiro
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renata S Fernandes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciano C Castro
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valbert N Cardoso
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mônica C Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Danyelle M Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, USA
| | - Alice Ferretti
- Department of Nuclear Medicine, Molecular Imaging, Radiology, Neuro Radiology, Medical Physics, Clinical Laboratory, Microbiology & Pathology, Santa Maria de la Misericordia Hospital, Rovigo, Italy
| | - Domenico Rubello
- Department of Nuclear Medicine, Molecular Imaging, Radiology, Neuro Radiology, Medical Physics, Clinical Laboratory, Microbiology & Pathology, Santa Maria de la Misericordia Hospital, Rovigo, Italy.
| | - Elaine A Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - André L B de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
67
|
Chandran P, Riviere JE, Monteiro-Riviere NA. Surface chemistry of gold nanoparticles determines the biocorona composition impacting cellular uptake, toxicity and gene expression profiles in human endothelial cells. Nanotoxicology 2017; 11:507-519. [PMID: 28420299 DOI: 10.1080/17435390.2017.1314036] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This study investigated the role of nanoparticle size and surface chemistry on biocorona composition and its effect on uptake, toxicity and cellular responses in human umbilical vein endothelial cells (HUVEC), employing 40 and 80 nm gold nanoparticles (AuNP) with branched polyethyleneimine (BPEI), lipoic acid (LA) and polyethylene glycol (PEG) coatings. Proteomic analysis identified 59 hard corona proteins among the various AuNP, revealing largely surface chemistry-dependent signature adsorbomes exhibiting human serum albumin (HSA) abundance. Size distribution analysis revealed the relative instability and aggregation inducing potential of bare and corona-bound BPEI-AuNP, over LA- and PEG-AuNP. Circular dichroism analysis showed surface chemistry-dependent conformational changes of proteins binding to AuNP. Time-dependent uptake of bare, plasma corona (PC) and HSA corona-bound AuNP (HSA-AuNP) showed significant reduction in uptake with PC formation. Cell viability studies demonstrated dose-dependent toxicity of BPEI-AuNP. Transcriptional profiling studies revealed 126 genes, from 13 biological pathways, to be differentially regulated by 40 nm bare and PC-bound BPEI-AuNP (PC-BPEI-AuNP). Furthermore, PC formation relieved the toxicity of cationic BPEI-AuNP by modulating expression of genes involved in DNA damage and repair, heat shock response, mitochondrial energy metabolism, oxidative stress and antioxidant response, and ER stress and unfolded protein response cascades, which were aberrantly expressed in bare BPEI-AuNP-treated cells. NP surface chemistry is shown to play the dominant role over size in determining the biocorona composition, which in turn modulates cell uptake, and biological responses, consequently defining the potential safety and efficacy of nanoformulations.
Collapse
Affiliation(s)
- Parwathy Chandran
- a Department of Anatomy and Physiology, Nanotechnology Innovation Center of Kansas State , Kansas State University , Manhattan , KS , USA
| | - Jim E Riviere
- b Department of Anatomy and Physiology, Institute of Computational Comparative Medicine , Kansas State University , Manhattan , KS , USA
| | - Nancy A Monteiro-Riviere
- a Department of Anatomy and Physiology, Nanotechnology Innovation Center of Kansas State , Kansas State University , Manhattan , KS , USA
| |
Collapse
|
68
|
Pederzoli F, Tosi G, Vandelli MA, Belletti D, Forni F, Ruozi B. Protein corona and nanoparticles: how can we investigate on? WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [DOI: 10.1002/wnan.1467] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 01/19/2017] [Accepted: 02/13/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Francesca Pederzoli
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| | - Giovanni Tosi
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| | | | - Daniela Belletti
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| | - Flavio Forni
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| | - Barbara Ruozi
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| |
Collapse
|
69
|
Liu L, Bi Y, Zhou M, Chen X, He X, Zhang Y, Sun T, Ruan C, Chen Q, Wang H, Jiang C. Biomimetic Human Serum Albumin Nanoparticle for Efficiently Targeting Therapy to Metastatic Breast Cancers. ACS APPLIED MATERIALS & INTERFACES 2017; 9:7424-7435. [PMID: 28150932 DOI: 10.1021/acsami.6b14390] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Triple-negative breast cancers (TNBCs), devoid of hormone receptors and human epidermal growth-factor receptor-2/Neu expression, bring about poor prognosis and induce a high rate of systematic metastases. The ineffectiveness of current therapies on TNBCs could be attributed to the lack of efficient targeted therapy. Paclitaxel (PTX) is considered one of first-line chemotherapeutics for TNBC treatment but, due to its low aqueous solubility and nonspecific accumulation, results in poor antitumor efficacy. The present study is aimed at enhancing the chemotherapeutic potency of PTX by improving the stability and targeting efficiency of PTX-loaded nanoparticulate drug carriers. Here, PTX was incorporated in nontoxic and endogenous material, human serum albumin (HSA), via an innovative disulfide reduction method to construct HSA-based PTX nanoparticle (HSA-PTX NP) to not only realize redox-responsive drug release but also improve in vivo stability. Besides, W peptide was selected as a target ligand to be conjugated with HSA-PTX NP for endowing active targeting ability. The resulting Wpep-HSA-PTX NP possessed a spherical structure (118 nm), 9.87% drug-loading content, and 86.3% entrapment efficiency. An in vitro drug release test showed that PTX release from Wpep-HSA-PTX NP was of a redox-responsive manner. Furthermore, cellular uptake of Wpep-HSA-PTX NP was significantly enhanced, exhibiting the improved antiproliferation and antitube formation effects of PTX in vitro. In comparison with those commercial formulations and conventional HSA NP, Wpep-HSA-PTX NP exhibited better pharmacokinetic behaviors and tumor homing characteristics. The antitumor efficacy of Wpep-HSA-PTX NP was further confirmed by the strong pro-apoptotic effect and reduced tumor burden. In a word, this evidence highlighted the proof of concept for Wpep-HSA NP as a promising conqueror to the ineffectiveness of TNBC therapy.
Collapse
Affiliation(s)
- Lisha Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
- National Pharmaceutical Engineering and Research Center, China State Institute of Pharmaceutical Industry , Shanghai 201203, China
| | - Yunke Bi
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
- Department of Neurosurgery, Shanghai First People's Hospital, Shanghai Jiao Tong University, School of Medicine , Shanghai 201620, China
| | - Muru Zhou
- National Pharmaceutical Engineering and Research Center, China State Institute of Pharmaceutical Industry , Shanghai 201203, China
| | - Xinli Chen
- National Pharmaceutical Engineering and Research Center, China State Institute of Pharmaceutical Industry , Shanghai 201203, China
| | - Xi He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Chunhui Ruan
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Qingjun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| | - Hao Wang
- National Pharmaceutical Engineering and Research Center, China State Institute of Pharmaceutical Industry , Shanghai 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University , 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
70
|
Oda CMR, Fernandes RS, de Araújo Lopes SC, de Oliveira MC, Cardoso VN, Santos DM, de Castro Pimenta AM, Malachias A, Paniago R, Townsend DM, Colletti PM, Rubello D, Alves RJ, de Barros ALB, Leite EA. Synthesis, characterization and radiolabeling of polymeric nano-micelles as a platform for tumor delivering. Biomed Pharmacother 2017; 89:268-275. [PMID: 28235689 DOI: 10.1016/j.biopha.2017.01.144] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/25/2017] [Indexed: 10/20/2022] Open
Abstract
The use of nanoparticles for diagnostic approaches leads to higher accumulation in the targeting tissue promoting a better signal-to-noise ratio and consequently, early tumor detection through scintigraphic techniques. Such approaches have inherent advantages, including the possibility of association with a variety of gamma-emitting radionuclides available, among them, Tecnethium-99m (99mTc). 99mTc is readily conjugated with nanoparticles using chelating agents, such as diethylenetriaminepentaacetic acid (DTPA). Leveraging this approach, we synthesized polymeric micelles (PM) consisting of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethyleneglycol)-2000] (DSPE-mPEG2000) functionalized with DTPA for radiolabeling with 99mTc. Micelles made up of DSPE-mPEG2000 and DSPE-PEG2000-DTPA had a mean diameter of ∼10nm, as measured by DLS and SAXS techniques, and a zeta potential of -2.7±1.1mV. Radiolabeled micelles exhibited high radiochemical yields and stability. In vivo assays indicated long blood circulation time (456.3min). High uptake in liver, spleen and kidneys was observed in the biodistribution and imaging studies on healthy and tumor-bearing mice. In addition, a high tumor-to-muscle ratio was detected, which increased over time, showing accumulation of the PM in the tumor region. These findings indicate that this system is a promising platform for simultaneous delivery of therapeutic agents and diagnostic probes.
Collapse
Affiliation(s)
- Caroline Mari Ramos Oda
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Renata Salgado Fernandes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Sávia Caldeira de Araújo Lopes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Mônica Cristina de Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Valbert Nascimento Cardoso
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Moreira Santos
- Department of Biochemistry and Imunology, Biological Science Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Department of Biochemistry and Imunology, Biological Science Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Angelo Malachias
- Department of Physics, Exact Sciences Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rogério Paniago
- Department of Physics, Exact Sciences Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Danyelle M Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, United States
| | - Patrick M Colletti
- Department of Nuclear Medicine, University of Southern California, Los Angeles, CA, United States
| | - Domenico Rubello
- Department of Nuclear Medicine, Imaging and Clinical Pathology, Santa Maria della Misericordia Hospital, Rovigo, Italy.
| | - Ricardo José Alves
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
71
|
Pisani C, Gaillard JC, Odorico M, Nyalosaso JL, Charnay C, Guari Y, Chopineau J, Devoisselle JM, Armengaud J, Prat O. The timeline of corona formation around silica nanocarriers highlights the role of the protein interactome. NANOSCALE 2017; 9:1840-1851. [PMID: 27858044 DOI: 10.1039/c6nr04765c] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Magnetic mesoporous silica nanoparticles (M-MSNs) represent promising targeting tools for theranostics. Engineering the interaction of nanoparticles (NPs) with biological systems requires an understanding of protein corona formation around the nanoparticles as this drives the biological fate of nanocarriers. We investigated the behavior of proteins in contact with M-MSNs by high-throughput comparative proteomics, using human and bovine sera as biological fluids, in order to assess the adsorption dynamics of proteins in these media. Using system biology tools, and especially protein-protein interaction databases, we demonstrated how the protein network builds up within the corona over the course of the experiment. Based on these results, we introduce and discuss the role of the "corona interactome" as an important factor influencing protein corona evolution. The concept of the "corona interactome" is an original methodology which could be generalized to all NP candidates. Based on this, pre-coating nanocarriers with specific proteins presenting minimal interactions with opsonins might provide them with properties such as stealth.
Collapse
Affiliation(s)
- Cédric Pisani
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France and CEA, Direction de la Recherche Fondamentale-BIAM, Site de Marcoule, F-30207 Bagnols-sur-Cèze, France.
| | - Jean-Charles Gaillard
- CEA, Direction de la Recherche Fondamentale-IBITECS, Site de Marcoule, F-30207 Bagnols-sur-Cèze, France
| | - Michaël Odorico
- Institut de Chimie Séparative de Marcoule, UMR 5257 CEA-CNRS-ENSCM-UM, Site de Marcoule, F-30207 Bagnols-sur-Cèze, France
| | - Jeff L Nyalosaso
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France
| | - Clarence Charnay
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France
| | - Yannick Guari
- Institut Charles Gerhardt de Montpellier, IMNO, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France
| | - Joël Chopineau
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France and Université de Nîmes Rue Georges Salan, F-30000 Nîmes, France
| | - Jean-Marie Devoisselle
- Institut Charles Gerhardt de Montpellier, MACS, UMR 5253 CNRS-ENSCM-UM, 1701, Place Eugène Bataillon, F-34095 Montpellier, France
| | - Jean Armengaud
- CEA, Direction de la Recherche Fondamentale-IBITECS, Site de Marcoule, F-30207 Bagnols-sur-Cèze, France
| | - Odette Prat
- CEA, Direction de la Recherche Fondamentale-BIAM, Site de Marcoule, F-30207 Bagnols-sur-Cèze, France.
| |
Collapse
|
72
|
Sobczynski DJ, Eniola-Adefeso O. Effect of anticoagulants on the protein corona-induced reduced drug carrier adhesion efficiency in human blood flow. Acta Biomater 2017; 48:186-194. [PMID: 27765678 PMCID: PMC5235944 DOI: 10.1016/j.actbio.2016.10.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 11/17/2022]
Abstract
Plasma proteins rapidly coat the surfaces of particulate drug carriers to form a protein corona upon their injection into the bloodstream. The high presence of immunoglobulins in the corona formed on poly(lactic-co-glycolic acid) (PLGA) vascular-targeted carrier (VTC) surfaces was recently shown to negatively impact their adhesion to activated endothelial cells (aECs) in vitro. Here, we characterized the influence of anticoagulants, or their absence, on the binding efficiency of VTCs of various materials via modulation of their protein corona. Specifically, we evaluated the adhesion of PLGA, poly(lactic acid) (PLA), polycaprolactone (PCL), silica, and polystyrene VTCs to aECs in heparinized, citrated, and non-anticoagulated (serum and whole) blood flows relative to buffer control. Particle adhesion is substantially reduced in non-anticoagulated blood flows regardless of the material type while only moderate to minimal reduction is observed for VTCs in anticoagulant-containing blood flow depending on the anticoagulant and material type. The substantial reduction in VTC adhesion in blood flows was linked to a high presence of immunoglobulin-sized proteins in the VTC corona via SDS-PAGE analysis. Of all the materials evaluated, PLGA was the most sensitive to plasma protein effects while PCL was the most resistant, suggesting particle hydrophobicity is a critical component of the observed negative plasma protein effects. Overall, this work demonstrates that anticoagulant positively alters the effect of plasma proteins in prescribing VTC adhesion to aECs in human blood flow, which has implication in the use of in vitro blood flow assays for functional evaluation of VTCs for in vivo use. STATEMENT OF SIGNIFICANCE This study addresses the impact of anticoagulant on altering the extent of the previously observed protein corona-induced adhesion reduction of vascular-targeted drug carriers in human blood flows. Specifically, serum blood flow (no anticoagulant) magnifies the negative effect of the plasma protein corona on drug carrier adhesion relative to citrated or heparinized blood flows. Overall, the results from this work suggest that serum better predicts targeted drug carrier adhesion efficiency in vivo compared to anticoagulant containing plasma. Furthermore, this study offers critical insight into the importance of how the choice of anticoagulant can greatly affect drug delivery-related processes in vitro.
Collapse
Affiliation(s)
- Daniel J Sobczynski
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 28109, United States.
| |
Collapse
|
73
|
McCarthy DP, Yap JWT, Harp CT, Song WK, Chen J, Pearson RM, Miller SD, Shea LD. An antigen-encapsulating nanoparticle platform for T H1/17 immune tolerance therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2017; 13:191-200. [PMID: 27720992 PMCID: PMC5237397 DOI: 10.1016/j.nano.2016.09.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/12/2016] [Accepted: 09/20/2016] [Indexed: 01/19/2023]
Abstract
Tolerogenic nanoparticles (NPs) are rapidly being developed as specific immunotherapies to treat autoimmune disease. However, many NP-based therapies conjugate antigen (Ag) directly to the NP posing safety concerns due to antibody binding or require the co-delivery of immunosuppressants to induce tolerance. Here, we developed Ag encapsulated NPs comprised of poly(lactide-co-glycolide) [PLG(Ag)] and investigated the mechanism of action for Ag-specific tolerance induction in an autoimmune model of T helper type 1/17 dysfunction - relapse-remitting experimental autoimmune encephalomyelitis (R-EAE). PLG(Ag) completely abrogated disease induction in an organ specific manner, where the spleen was dispensable for tolerance induction. PLG(Ag) delivered intravenously distributed to the liver, associated with macrophages, and recruited Ag-specific T cells. Furthermore, programmed death ligand 1 (PD-L1) was increased on Ag presenting cells and PD-1 blockade lessened tolerance induction. The robust promotion of tolerance by PLG(Ag) without co-delivery of immunosuppressive drugs, suggests that these NPs effectively deliver antigen to endogenous tolerogenic pathways.
Collapse
Affiliation(s)
- Derrick P McCarthy
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Christopher T Harp
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - W Kelsey Song
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Jeane Chen
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, Evanston, IL, USA; Department of Chemical and Biological Engineering, Evanston, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
74
|
Némethová V, Buliaková B, Mazancová P, Bábelová A, Šelc M, Moravčíková D, Kleščíková L, Ursínyová M, Gábelová A, Rázga F. Intracellular uptake of magnetite nanoparticles: A focus on physico-chemical characterization and interpretation of in vitro data. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:161-168. [DOI: 10.1016/j.msec.2016.08.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/18/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
|
75
|
Choi K, Riviere JE, Monteiro-Riviere NA. Protein corona modulation of hepatocyte uptake and molecular mechanisms of gold nanoparticle toxicity. Nanotoxicology 2016; 11:64-75. [DOI: 10.1080/17435390.2016.1264638] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Kyoungju Choi
- Department of Anatomy and Physiology, Kansas State University, Nanotechnology Innovation Center of Kansas State (NICKS), Manhattan, KS, USA
| | - Jim E. Riviere
- Department of Anatomy and Physiology, Kansas State University, Nanotechnology Innovation Center of Kansas State (NICKS), Manhattan, KS, USA
| | - Nancy A. Monteiro-Riviere
- Department of Anatomy and Physiology, Kansas State University, Nanotechnology Innovation Center of Kansas State (NICKS), Manhattan, KS, USA
| |
Collapse
|
76
|
Yousefi N, Tufenkji N. Probing the Interaction between Nanoparticles and Lipid Membranes by Quartz Crystal Microbalance with Dissipation Monitoring. Front Chem 2016; 4:46. [PMID: 27995125 PMCID: PMC5136538 DOI: 10.3389/fchem.2016.00046] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/18/2016] [Indexed: 11/13/2022] Open
Abstract
There is increasing interest in using quartz crystal microbalance with dissipation monitoring (QCM-D) to investigate the interaction of nanoparticles (NPs) with model surfaces. The high sensitivity, ease of use and the ability to monitor interactions in real-time has made it a popular technique for colloid chemists, biologists, bioengineers, and biophysicists. QCM-D has been recently used to probe the interaction of NPs with supported lipid bilayers (SLBs) as model cell membranes. The interaction of NPs with SLBs is highly influenced by the quality of the lipid bilayers. Unlike many surface sensitive techniques, by using QCM-D, the quality of SLBs can be assessed in real-time, hence QCM-D studies on SLB-NP interactions are less prone to the artifacts arising from bilayers that are not well formed. The ease of use and commercial availability of a wide range of sensor surfaces also have made QCM-D a versatile tool for studying NP interactions with lipid bilayers. In this review, we summarize the state-of-the-art on QCM-D based techniques for probing the interactions of NPs with lipid bilayers.
Collapse
Affiliation(s)
- Nariman Yousefi
- Department of Chemical Engineering, McGill University Montreal, QC, Canada
| | - Nathalie Tufenkji
- Department of Chemical Engineering, McGill University Montreal, QC, Canada
| |
Collapse
|
77
|
Müller J, Bauer KN, Prozeller D, Simon J, Mailänder V, Wurm FR, Winzen S, Landfester K. Coating nanoparticles with tunable surfactants facilitates control over the protein corona. Biomaterials 2016; 115:1-8. [PMID: 27871002 DOI: 10.1016/j.biomaterials.2016.11.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/10/2016] [Accepted: 11/13/2016] [Indexed: 12/18/2022]
Abstract
Nanoparticles with long blood circulation time are a prerequisite for targeted drug delivery. To make the nanoparticles invisible for phagocytizing cells, functional moieties on the particle surface are believed to be necessary to attract specific so-called 'stealth' proteins forming a protein 'corona'. Currently, covalent attachment of those moieties represents the only way to achieve that attraction. However, that approach requires a high synthetic effort and is difficult to control. Therefore, we present the coating of model nanoparticles with biodegradable polymeric surfactants as an alternative method. The thermodynamic parameters of the coating process can be tuned by adjusting the surfactants' block lengths and hydrophilicity. Consequently, the unspecific protein adsorption and aggregation tendency of the particles can be controlled, and stealth proteins inhibiting cell uptake are enriched on their surface. This non-covalent approach could be applied to any particle type and thus facilitates tuning the protein corona and its biological impact.
Collapse
Affiliation(s)
- J Müller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - K N Bauer
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - D Prozeller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - J Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - V Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Dermatology Clinic, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - F R Wurm
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - S Winzen
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - K Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
78
|
Bugno J, Hsu HJ, Hong S. Tweaking dendrimers and dendritic nanoparticles for controlled nano-bio interactions: potential nanocarriers for improved cancer targeting. J Drug Target 2016; 23:642-50. [PMID: 26453160 DOI: 10.3109/1061186x.2015.1052077] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanoparticles have shown great promise in the treatment of cancer, with a demonstrated potential in targeted drug delivery. Among a myriad of nanocarriers that have been recently developed, dendrimers have attracted a great deal of scientific interests due to their unique chemical and structural properties that allow for precise engineering of their characteristics. Despite this, the clinical translation of dendrimers has been hindered due to their drawbacks, such as scale-up issues, rapid systemic elimination, inefficient tumor accumulation and limited drug loading. In order to overcome these limitations, a series of reengineered dendrimers have been recently introduced using various approaches, including: (i) modifications of structure and surfaces; (ii) integration with linear polymers and (iii) hybridization with other types of nanocarriers. Chemical modifications and surface engineering have tailored dendrimers to improve their pharmacokinetics and tissue permeation. Copolymerization of dendritic polymers with linear polymers has resulted in various amphiphilic copolymers with self-assembly capabilities and improved drug loading efficiencies. Hybridization with other nanocarriers integrates advantageous characteristics of both systems, which includes prolonged plasma circulation times and enhanced tumor targeting. This review provides a comprehensive summary of the newly emerging drug delivery systems that involve reengineering of dendrimers in an effort to precisely control their nano-bio interactions, mitigating their inherent weaknesses.
Collapse
Affiliation(s)
- Jason Bugno
- a Department of Biopharmaceutical Sciences , College of Pharmacy, University of Illinois , Chicago , IL , USA and
| | - Hao-Jui Hsu
- a Department of Biopharmaceutical Sciences , College of Pharmacy, University of Illinois , Chicago , IL , USA and
| | - Seungpyo Hong
- a Department of Biopharmaceutical Sciences , College of Pharmacy, University of Illinois , Chicago , IL , USA and.,b Integrated Science and Engineering Division, Underwood International College, Yonsei University , Seoul , Korea
| |
Collapse
|
79
|
Pearson RM, Sen S, Hsu HJ, Pasko M, Gaske M, Král P, Hong S. Tuning the Selectivity of Dendron Micelles Through Variations of the Poly(ethylene glycol) Corona. ACS NANO 2016; 10:6905-6914. [PMID: 27267700 PMCID: PMC6800011 DOI: 10.1021/acsnano.6b02708] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Engineering controllable cellular interactions into nanoscale drug delivery systems is key to enable their full potential. Here, using folic acid (FA) as a model targeting ligand and dendron micelles (DM) as a nanoparticle (NP) platform, we present a comprehensive experimental and modeling investigation of the structural properties of DMs that govern the formation of controllable, FA-mediated cellular interactions. Our experimental results demonstrate that a high level of control over the specific cell interactions of FA-targeted DMs can be achieved through modulation of the PEG corona length and the FA content. Using various molecular weight PEGs (0.6K, 1K, and 2K g/mol) and contents of dendron-FA conjugate incorporated into DMs (0, 5, 10, 25 wt %), the cell interactions of the targeted DMs could be controlled to exhibit minimal to >25-fold enhancement over nontargeted DMs. Molecular dynamics simulations indicated that structural characteristics, such as solvent accessible surface area of FA, local PEG density near FA, and FA mobility, account in part for the experimental differences in cellular interactions. The molecular structure that allows FA to depart from the surface of DMs to facilitate the initial cell surface binding was revealed to be the most important contributor for determining FA-mediated cellular interactions of DMs. The modular properties of DMs in controlling their specific cell interactions support the potential of DMs as a delivery platform and offer design cues for future development of targeted NPs.
Collapse
Affiliation(s)
- Ryan M. Pearson
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| | - Soumyo Sen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hao-jui Hsu
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| | - Matt Pasko
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| | - Marilyn Gaske
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Physics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Seungpyo Hong
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Integrated OMICs for Biomedical Science, Yonsei University, Seoul, 03706, Republic of Korea
- Underwood International College, Yonsei University, Seoul, 03706, Republic of Korea
| |
Collapse
|
80
|
Zhong Q, Bielski ER, Rodrigues LS, Brown MR, Reineke JJ, da Rocha SRP. Conjugation to Poly(amidoamine) Dendrimers and Pulmonary Delivery Reduce Cardiac Accumulation and Enhance Antitumor Activity of Doxorubicin in Lung Metastasis. Mol Pharm 2016; 13:2363-75. [PMID: 27253493 PMCID: PMC6886243 DOI: 10.1021/acs.molpharmaceut.6b00126] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lung is one of the most common sites to which almost all other primary tumors metastasize. The major challenges in the chemotherapy of lung metastases include the low drug concentration found in the tumors and high systemic toxicity upon systemic administration. In this study, we combine local lung delivery and the use of nanocarrier-based systems for improving pharmacokinetics and biodistribution of the therapeutics to fight lung metastases. We investigate the impact of the conjugation of doxorubicin (DOX) to carboxyl-terminated poly(amidoamine) dendrimers (PAMAM) through a bond that allows for intracellular-triggered release, and the effect of pulmonary delivery of the dendrimer-DOX conjugate in decreasing tumor burden in a lung metastasis model. The results show a dramatic increase in efficacy of DOX treatment of the melanoma (B16-F10) lung metastasis mouse model upon pulmonary administration of the drug, as indicated by decreased tumor burden (lung weight) and increased survival rates of the animals (male C57BL/6) when compared to iv delivery. Conjugation of DOX further increased the therapeutic efficacy upon lung delivery as indicated by the smaller number of nodules observed in the lungs when compared to free DOX. These results are in agreement with the biodistribution characteristics of the DOX upon pulmonary delivery, which showed a longer lung accumulation/retention compared to iv administration. The distribution of DOX to the heart tissue is also significantly decreased upon pulmonary administration, and further decreased upon conjugation. The results show, therefore, that pulmonary administration of DOX combined to conjugation to PAMAM dendrimer through an intracellular labile bond is a potential strategy to enhance the therapeutic efficacy and decrease systemic toxicity of DOX.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | - Elizabeth R. Bielski
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | - Leonan S. Rodrigues
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | - Matthew R. Brown
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | - Joshua J. Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Sandro R. P. da Rocha
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
- Department of Pharmaceutics, College of Pharmacy, and Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
81
|
Toy R, Roy K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng Transl Med 2016; 1:47-62. [PMID: 29313006 PMCID: PMC5689503 DOI: 10.1002/btm2.10005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022] Open
Abstract
Advances in immunotherapy have led to the development of a variety of promising therapeutics, including small molecules, proteins and peptides, monoclonal antibodies, and cellular therapies. Despite this wealth of new therapeutics, the efficacy of immunotherapy has been limited by challenges in targeted delivery and controlled release, that is, spatial and temporal control on delivery. Particulate carriers, especially nanoparticles have been widely studied in drug delivery and vaccine research and are being increasingly investigated as vehicles to deliver immunotherapies. Nanoparticle-mediated drug delivery could provide several benefits, including control of biodistribution and transport kinetics, the potential for site-specific targeting, immunogenicity, tracking capability using medical imaging, and multitherapeutic loading. There are also a unique set of challenges, which include nonspecific uptake by phagocytic cells, off-target biodistribution, permeation through tissue (transport limitation), nonspecific immune-activation, and poor control over intracellular localization. This review highlights the importance of understanding the relationship between a nanoparticle's size, shape, charge, ligand density and elasticity to its vascular transport, biodistribution, cellular internalization, and immunogenicity. For the design of an effective immunotherapy, we highlight the importance of selecting a nanoparticle's physical characteristics (e.g., size, shape, elasticity) and its surface functionalization (e.g., chemical or polymer modifications, targeting or tissue-penetrating peptides) with consideration of its reactivity to the targeted microenvironment (e.g., targeted cell types, use of stimuli-sensitive biomaterials, immunogenicity). Applications of this rational nanoparticle design process in vaccine development and cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Randall Toy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| | - Krishnendu Roy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| |
Collapse
|
82
|
Zhang Y, Buhrman JS, Liu Y, Rayahin JE, Gemeinhart RA. Reducible Micelleplexes are Stable Systems for Anti-miRNA Delivery in Cerebrospinal Fluid. Mol Pharm 2016; 13:1791-9. [PMID: 27177352 DOI: 10.1021/acs.molpharmaceut.5b00933] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) and other central nervous system (CNS) cancers have poor long-term prognosis, and there is a significant need for improved treatments. GBM initiation and progression are mediated, in part, by microRNA (miRNA), which are endogenous posttranscriptional gene regulators. Misregulation of miRNAs is a potential target for therapeutic intervention in GBM. In this work, a micelle-like nanoparticle delivery system based upon the block copolymer poly(ethylene glycol-b-lactide-b-arginine) was designed with and without a reducible linkage between the lactide and RNA-binding peptide, R15, to assess the ability of the micelle-like particles to disassemble. Using confocal live cell imaging, intracellular dissociation was pronounced for the reducible micelleplexes. This dissociation was also supported by higher efficiency in a dual luciferase assay specific for the miRNA of interest, miR-21. Notably, micelleplexes were found to have significantly better stability and higher anti-miRNA activity in cerebrospinal fluid than in human plasma, suggesting an advantage for applying micelleplexes to CNS diseases and in vivo CNS therapeutics. The reducible delivery system was determined to be a promising delivery platform for the treatment of CNS diseases with miRNA therapy.
Collapse
Affiliation(s)
| | | | | | | | - Richard A Gemeinhart
- Department of Bioengineering, University of Illinois , Chicago, Illinois 60607-7052, United States
| |
Collapse
|
83
|
Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chem Rev 2016; 116:2602-63. [PMID: 26854975 PMCID: PMC5509216 DOI: 10.1021/acs.chemrev.5b00346] [Citation(s) in RCA: 1708] [Impact Index Per Article: 189.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Basit Yameen
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
84
|
Palchetti S, Pozzi D, Mahmoudi M, Caracciolo G. Exploitation of nanoparticle–protein corona for emerging therapeutic and diagnostic applications. J Mater Chem B 2016; 4:4376-4381. [DOI: 10.1039/c6tb01095d] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exposure of nanoparticles (NPs) to biological fluids (e.g., plasma, interstitial fluid, and cytoplasm) leads to the absorption of proteins on the NP surface, forming a protein corona (PC) that drastically influences the NP physicochemical properties.
Collapse
Affiliation(s)
- S. Palchetti
- Department of Molecular Medicine
- Rome
- Italy
- Istituti Fisioterapici Ospitalieri
- Istituto Regina Elena
| | - D. Pozzi
- Department of Molecular Medicine
- Rome
- Italy
- Istituti Fisioterapici Ospitalieri
- Istituto Regina Elena
| | - M. Mahmoudi
- Department of Nanotechnology and Nanotechnology Research Center
- Faculty of Pharmacy
- Tehran University of Medical Sciences
- Tehran 13169-43551
- Iran
| | - G. Caracciolo
- Department of Molecular Medicine
- Rome
- Italy
- Istituti Fisioterapici Ospitalieri
- Istituto Regina Elena
| |
Collapse
|
85
|
Vasti C, Bedoya DA, Rojas R, Giacomelli CE. Effect of the protein corona on the colloidal stability and reactivity of LDH-based nanocarriers. J Mater Chem B 2016; 4:2008-2016. [DOI: 10.1039/c5tb02698a] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The physicochemical properties of drug nanocarriers such as layered double hydroxides nanoparticles (LDH-NPs) determine their circulation times in biological media and their interaction with the targeted cells.
Collapse
Affiliation(s)
- Cecilia Vasti
- INFIQC-CONICET
- Departamento de Fisicoquímica
- Facultad de Ciencias Químicas
- Universidad Nacional de Córdoba
- X5000HUA Córdoba
| | - Dariana Aristizabal Bedoya
- INFIQC-CONICET
- Departamento de Fisicoquímica
- Facultad de Ciencias Químicas
- Universidad Nacional de Córdoba
- X5000HUA Córdoba
| | - Ricardo Rojas
- INFIQC-CONICET
- Departamento de Fisicoquímica
- Facultad de Ciencias Químicas
- Universidad Nacional de Córdoba
- X5000HUA Córdoba
| | - Carla E. Giacomelli
- INFIQC-CONICET
- Departamento de Fisicoquímica
- Facultad de Ciencias Químicas
- Universidad Nacional de Córdoba
- X5000HUA Córdoba
| |
Collapse
|
86
|
Namdee K, Sobczynski DJ, Onyskiw PJ, Eniola-Adefeso O. Differential Impact of Plasma Proteins on the Adhesion Efficiency of Vascular-Targeted Carriers (VTCs) in Blood of Common Laboratory Animals. Bioconjug Chem 2015; 26:2419-28. [PMID: 26505780 PMCID: PMC4866610 DOI: 10.1021/acs.bioconjchem.5b00474] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vascular-targeted carrier (VTC) interaction with human plasma is known to reduce targeted adhesion efficiency in vitro. However, the role of plasma proteins on the adhesion efficiency of VTCs in laboratory animals remains unknown. Here, in vitro blood flow assays are used to explore the effects of plasma from mouse, rabbit, and porcine on VTC adhesion. Porcine blood exhibited a strong negative plasma effect on VTC adhesion while no significant plasma effect was found with rabbit and mouse blood. A brush density poly(ethylene glycol) (PEG) on VTCs was effective at improving adhesion of microsized, but not nanosized, VTCs in porcine blood. Overall, the results suggest that porcine models, as opposed to mouse, can serve as better models in preclinical research for predicting the in vivo functionality of VTCs for use in humans. These considerations hold great importance for the design of various pharmaceutical products and development of reliable drug delivery systems.
Collapse
Affiliation(s)
| | | | - Peter J. Onyskiw
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| |
Collapse
|
87
|
Weidner A, Gräfe C, von der Lühe M, Remmer H, Clement JH, Eberbeck D, Ludwig F, Müller R, Schacher FH, Dutz S. Preparation of Core-Shell Hybrid Materials by Producing a Protein Corona Around Magnetic Nanoparticles. NANOSCALE RESEARCH LETTERS 2015; 10:992. [PMID: 26153125 PMCID: PMC4495093 DOI: 10.1186/s11671-015-0992-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/24/2015] [Indexed: 05/18/2023]
Abstract
Nanoparticles experience increasing interest for a variety of medical and pharmaceutical applications. When exposing nanomaterials, e.g., magnetic iron oxide nanoparticles (MNP), to human blood, a protein corona consisting of various components is formed immediately. The composition of the corona as well as its amount bound to the particle surface is dependent on different factors, e.g., particle size and surface charge. The actual composition of the formed protein corona might be of major importance for cellular uptake of magnetic nanoparticles. The aim of the present study was to analyze the formation of the protein corona during in vitro serum incubation in dependency of incubation time and temperature. For this, MNP with different shells were incubated in fetal calf serum (FCS, serving as protein source) within a water bath for a defined time and at a defined temperature. Before and after incubation the particles were characterized by a variety of methods. It was found that immediately (seconds) after contact of MNP and FCS, a protein corona is formed on the surface of MNP. This formation led to an increase of particle size and a slight agglomeration of the particles, which was relatively constant during the first minutes of incubation. A longer incubation (from hours to days) resulted in a stronger agglomeration of the FCS incubated MNP. Quantitative analysis (gel electrophoresis) of serum-incubated particles revealed a relatively constant amount of bound proteins during the first minutes of serum incubation. After a longer incubation (>20 min), a considerably higher amount of surface proteins was determined for incubation temperatures below 40 °C. For incubation temperatures above 50 °C, the influence of time was less significant which might be attributed to denaturation of proteins during incubation. Overall, analysis of the molecular weight distribution of proteins found in the corona revealed a clear influence of incubation time and temperature on corona composition.
Collapse
Affiliation(s)
- A Weidner
- />Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau, Gustav-Kirchhoff-Straße 2, 98693 Ilmenau, Germany
| | - C Gräfe
- />Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Erlanger Allee 101, 07747 Jena, Germany
| | - M von der Lühe
- />Institute of Organic and Macromolecular Chemistry, Friedrich-Schiller-University Jena, Humboldtstraße 10, 07743 Jena, Germany
- />Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - H Remmer
- />Institut für Elektrische Messtechnik und Grundlagen der Elektrotechnik, Technische Universität Braunschweig, Hans-Sommer-Straße 66, 38106 Braunschweig, Germany
| | - J H Clement
- />Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Erlanger Allee 101, 07747 Jena, Germany
- />Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - D Eberbeck
- />Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - F Ludwig
- />Institut für Elektrische Messtechnik und Grundlagen der Elektrotechnik, Technische Universität Braunschweig, Hans-Sommer-Straße 66, 38106 Braunschweig, Germany
| | - R Müller
- />Department of Nano Biophotonics, Leibniz Institute of Photonic Technology (IPHT), A.-Einstein-Straße 9, 07745 Jena, Germany
| | - F H Schacher
- />Institute of Organic and Macromolecular Chemistry, Friedrich-Schiller-University Jena, Humboldtstraße 10, 07743 Jena, Germany
- />Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - S Dutz
- />Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau, Gustav-Kirchhoff-Straße 2, 98693 Ilmenau, Germany
- />Department of Nano Biophotonics, Leibniz Institute of Photonic Technology (IPHT), A.-Einstein-Straße 9, 07745 Jena, Germany
| |
Collapse
|
88
|
Shetab Boushehri MA, Lamprecht A. Nanoparticles as drug carriers: current issues with in vitro testing. Nanomedicine (Lond) 2015; 10:3213-30. [DOI: 10.2217/nnm.15.154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Incorporation of nanotechnology in the field of drug delivery has created exciting opportunities for the purposeful design of nanocarriers with potentials such as targeted delivery or controlled release of the incorporated cargo, improvement of bioavailability and reduction of therapeutic side-effects. Prior to in vivo administration, nanocarriers should undergo a set of in vitro evaluation procedures to ensure their stability, safety, conformity and ability to fulfill the desired mission. In this paper, current issues with in vitro evaluation techniques used for nanocarrier characterization (assessment of particle size, surface charge, drug release and toxicity) will be discussed. Furthermore, sufficiency of in vitro evaluation procedures for the prediction of in vivo scenarios and the necessary considerations to improve the correlation between the two settings will be debated.
Collapse
Affiliation(s)
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
- Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France
| |
Collapse
|
89
|
Caracciolo G, Palchetti S, Colapicchioni V, Digiacomo L, Pozzi D, Capriotti AL, La Barbera G, Laganà A. Stealth effect of biomolecular corona on nanoparticle uptake by immune cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:10764-73. [PMID: 26378619 DOI: 10.1021/acs.langmuir.5b02158] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
When injected in a biological milieu, a nanomaterial rapidly adsorbs biomolecules forming a biomolecular corona. The biomolecular corona changes the interfacial composition of a nanomaterial giving it a biological identity that determines the physiological response. Characterization of the biomolecular structure and composition has received increasing attention mostly due to its detrimental impact on the nanomaterial's metabolism in vivo. It is generally accepted that an opsonin-enriched biomolecular corona promotes immune system recognition and rapid clearance from circulation. Here we applied dynamic light scattering and nanoliquid chromatography tandem mass spectrometry to thoroughly characterize the biomolecular corona formed around lipid and silica nanoparticles (NPs). Incubation with human plasma resulted in the formation of NP-biomolecular coronas enriched with immunoglobulins, complement factors, and coagulation proteins that bind to surface receptors on immune cells and elicit phagocytosis. Conversely, we found that protein-coated NPs were protected from uptake by macrophage RAW 264.7 cells. This implies that the biomolecular corona formation provides a stealth effect on macrophage recognition. Our results suggest that correct prediction of the NP's fate in vivo will require more than just the knowledge of the biomolecular corona composition. Validation of efficient methods for mapping protein binding sites on the biomolecular corona of NPs is an urgent task for future research.
Collapse
Affiliation(s)
- Giulio Caracciolo
- Department of Molecular Medicine, "Sapienza" University of Rome , Viale Regina Elena 291, 00161 Rome, Italy
| | - Sara Palchetti
- Department of Molecular Medicine, "Sapienza" University of Rome , Viale Regina Elena 291, 00161 Rome, Italy
| | - Valentina Colapicchioni
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia Viale Regina Elena 291, 00161 Roma, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, "Sapienza" University of Rome , Viale Regina Elena 291, 00161 Rome, Italy
- Department of Bioscience and Biotechnology, University of Camerino , Via Gentile III da Varano, Camerino, Province of Macerata 62032, Italy
| | - Daniela Pozzi
- Department of Molecular Medicine, "Sapienza" University of Rome , Viale Regina Elena 291, 00161 Rome, Italy
| | - Anna Laura Capriotti
- Department of Chemistry, "Sapienza" University of Rome , P.le A. Moro 5, 00185 Rome, Italy
| | - Giorgia La Barbera
- Department of Chemistry, "Sapienza" University of Rome , P.le A. Moro 5, 00185 Rome, Italy
| | - Aldo Laganà
- Department of Chemistry, "Sapienza" University of Rome , P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
90
|
Schneider CS, Bhargav AG, Perez JG, Wadajkar AS, Winkles JA, Woodworth GF, Kim AJ. Surface plasmon resonance as a high throughput method to evaluate specific and non-specific binding of nanotherapeutics. J Control Release 2015; 219:331-344. [PMID: 26415854 DOI: 10.1016/j.jconrel.2015.09.048] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/22/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022]
Abstract
Surface plasmon resonance (SPR) is a powerful analytical technique used to quantitatively examine the interactions between various biomolecules, such as proteins and nucleic acids. The technique has been particularly useful in screening and evaluating binding affinity of novel small molecule and biomolecule-derived therapeutics for various diseases and applications including lupus medications, thrombin inhibitors, HIV protease inhibitors, DNA gyrase inhibitors and many others. Recently, there has been increasing interest in nanotherapeutics (nanoRx), due to their unique properties and potential for controlled release of encapsulated drugs and structure-specific targeting to diseased tissues. NanoRx offer the potential to solve many drug delivery challenges by enabling, specific interactions between molecules on the surface of the nanoparticle and molecules in the diseased tissue, while minimizing off-target interactions toward non-diseased tissues. These properties are largely dependent upon careful control and balance of nanoRx interactions and binding properties with tissues in vivo. Given the great promise of nanoRx with regard to engineering specific molecular interactions, SPR can rapidly quantify small aliquots of nanoRx formulations for desired and undesired molecular interactions. Moving forward, we believe that utilization of SPR in the screening and design of nanoRx has the potential to greatly improve the development of targeted nanoRx formulations and eventually lead to improved therapeutic efficacy. In this review, we discuss (1) the fundamental principles of SPR and basic quantitative analysis of SPR data, (2) previous applications of SPR in the study of non-particulate therapeutics and nanoRx, and (3) future opportunities for the use of SPR in the evaluation of nanoRx.
Collapse
Affiliation(s)
- Craig S Schneider
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Adip G Bhargav
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jimena G Perez
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aniket S Wadajkar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A Winkles
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
91
|
Silver nanoparticle protein corona and toxicity: a mini-review. J Nanobiotechnology 2015; 13:55. [PMID: 26337542 PMCID: PMC4559354 DOI: 10.1186/s12951-015-0114-4] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/08/2015] [Indexed: 12/31/2022] Open
Abstract
Silver nanoparticles are one of the most important materials in the nanotechnology industry. Additionally, the protein corona is emerging as a key entity at the nanobiointerface; thus, a comprehensive understanding of the interactions between proteins and silver nanoparticles is imperative. Therefore, literature reporting studies involving both single molecule protein coronas (i.e., bovine and human serum albumin, tubulin, ubiquitin and hyaluronic-binding protein) and complex protein coronas (i.e., fetal bovine serum and yeast extract proteins) were selected to demonstrate the effects of protein coronas on silver nanoparticle cytotoxicity and antimicrobial activity. There is evidence that distinct and differential protein components may yield a "protein corona signature" that is related to the size and/or surface curvature of the silver nanoparticles. Therefore, the formation of silver nanoparticle protein coronas together with the biological response to these coronas (i.e., oxidative stress, inflammation and cytotoxicity) as well as other cellular biophysicochemical mechanisms (i.e., endocytosis, biotransformation and biodistribution) will be important for nanomedicine and nanotoxicology. Researchers may benefit from the information contained herein to improve biotechnological applications of silver nanoparticles and to address related safety concerns. In summary, the main aim of this mini-review is to highlight the relationship between the formation of silver nanoparticle protein coronas and toxicity.
Collapse
|
92
|
Piñeiro Y, Vargas Z, Rivas J, López-Quintela MA. Iron Oxide Based Nanoparticles for Magnetic Hyperthermia Strategies in Biological Applications. Eur J Inorg Chem 2015. [DOI: 10.1002/ejic.201500598] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
93
|
Zhang Y, Liu Y, Sen S, Král P, Gemeinhart RA. Charged group surface accessibility determines micelleplexes formation and cellular interaction. NANOSCALE 2015; 7:7559-7564. [PMID: 25866141 PMCID: PMC4479253 DOI: 10.1039/c5nr00095e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Micelleplexes are a class of nucleic acid carriers that have gained acceptance due to their size, stability, and ability to synergistically carry small molecules. MicroRNAs (miRNAs) are small non-coding RNA gene regulator that is consists of 19-22 nucleotides. Altered expression of miRNAs plays an important role in many human diseases. Using a model 22-nucleotide miRNA sequence, we investigated the interaction between charged groups on the micelle surface and miRNA. The model micelle system was formed from methoxy-poly(ethylene glycol)-b-poly(lactide) (mPEG-PLA) mixed with methoxy-poly(ethylene glycol)-b-poly(lactide)-b-oligoarginine (mPEG-PLA-Rx, x = 8 or 15). Surface properties of the micelles were varied by controlling the oligoarginine block length and conjugation density. Micelles were observed to have a core-shell conformation in the aqueous environment where the PLA block constituted the hydrophobic core, mPEG and oligoarginine formed a hydrophilic corona. Significantly different thermodynamic behaviors were observed during the interaction of single stranded miRNA with micelles of different surface properties, and the resulting micelleplexes mediated substantial cellular association. Depending upon the oligoarginine length and density, micelles exhibited miRNA loading capacity directly related to the presentation of charged groups on the surface. The effect of charged group accessibility of cationic micelle on micelleplex properties provides guidance on future miRNA delivery system design.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biopharmaceutical Sciences, University of Illinois, Chicago, IL 60612, USA.
| | | | | | | | | |
Collapse
|
94
|
Salatin S, Maleki Dizaj S, Yari Khosroushahi A. Effect of the surface modification, size, and shape on cellular uptake of nanoparticles. Cell Biol Int 2015; 39:881-90. [DOI: 10.1002/cbin.10459] [Citation(s) in RCA: 307] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/06/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Sara Salatin
- Biotechnology Research Center, Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
- Student Research Committee; Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
- Department of Pharmaceutical Nanotechnology; Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
| | - Solmaz Maleki Dizaj
- Biotechnology Research Center, Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
- Student Research Committee; Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
- Department of Pharmaceutical Nanotechnology; Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center; Faculty of Pharmacy; Tabriz University of Medical Science; Tabriz Iran
- Department of Pharmacognosy; Faculty of Pharmacy; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|