1
|
Zheng JJ, Li QZ, Wang Z, Wang X, Zhao Y, Gao X. Computer-aided nanodrug discovery: recent progress and future prospects. Chem Soc Rev 2024; 53:9059-9132. [PMID: 39148378 DOI: 10.1039/d3cs00575e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Nanodrugs, which utilise nanomaterials in disease prevention and therapy, have attracted considerable interest since their initial conceptualisation in the 1990s. Substantial efforts have been made to develop nanodrugs for overcoming the limitations of conventional drugs, such as low targeting efficacy, high dosage and toxicity, and potential drug resistance. Despite the significant progress that has been made in nanodrug discovery, the precise design or screening of nanomaterials with desired biomedical functions prior to experimentation remains a significant challenge. This is particularly the case with regard to personalised precision nanodrugs, which require the simultaneous optimisation of the structures, compositions, and surface functionalities of nanodrugs. The development of powerful computer clusters and algorithms has made it possible to overcome this challenge through in silico methods, which provide a comprehensive understanding of the medical functions of nanodrugs in relation to their physicochemical properties. In addition, machine learning techniques have been widely employed in nanodrug research, significantly accelerating the understanding of bio-nano interactions and the development of nanodrugs. This review will present a summary of the computational advances in nanodrug discovery, focusing on the understanding of how the key interfacial interactions, namely, surface adsorption, supramolecular recognition, surface catalysis, and chemical conversion, affect the therapeutic efficacy of nanodrugs. Furthermore, this review will discuss the challenges and opportunities in computer-aided nanodrug discovery, with particular emphasis on the integrated "computation + machine learning + experimentation" strategy that can potentially accelerate the discovery of precision nanodrugs.
Collapse
Affiliation(s)
- Jia-Jia Zheng
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Qiao-Zhi Li
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Zhenzhen Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Xiaoli Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yuliang Zhao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Xingfa Gao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
2
|
Yu T, Wang J, Zhou Y, Ma C, Bai R, Huang C, Wang S, Liu K, Han B. Harnessing Engineered Extracellular Vesicles from Mesenchymal Stem Cells as Therapeutic Scaffolds for Bone‐Related Diseases. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202402861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Indexed: 10/05/2024]
Abstract
AbstractMesenchymal stem cells (MSCs) play a crucial role in maintaining bone homeostasis and are extensively explored for cell therapy in various bone‐related diseases. In addition to direct cell therapy, the secretion of extracellular vesicles (EVs) by MSCs has emerged as a promising alternative approach. MSC‐derived EVs (MSC‐EVs) offer equivalent therapeutic efficacy to MSCs while mitigating potential risks. These EVs possess unique properties that enable them to traverse biological barriers and deliver bioactive cargos to target cells. Furthermore, by employing modification and engineering strategies, the therapeutic effects and tissue targeting specificity of MSC‐EVs can be further enhanced to meet specific therapeutic needs. In this review, the mechanisms and advantages of MSC‐EV therapy in diseased bone tissues are highlighted. Through simple isolation and modification techniques, MSC‐EV‐based biomaterials have demonstrated great promise for bone regeneration. Finally, future perspectives on MSC‐EV therapy are presented, envisioning the development of next‐generation regenerative materials and bioactive agents for clinical translation in the field of bone regeneration.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Jingwei Wang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Yusai Zhou
- School of Materials Science and Engineering Beihang University Beijing 100191 P. R. China
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Rushui Bai
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Cancan Huang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Shidong Wang
- Musculoskeletal Tumor Center Peking University People's Hospital No.11 Xizhimen South St. Beijing 100044 P. R. China
| | - Kai Liu
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Bing Han
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| |
Collapse
|
3
|
Chen LH, Hu JN. Development of nano-delivery systems for loaded bioactive compounds: using molecular dynamics simulations. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 38206576 DOI: 10.1080/10408398.2023.2301427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Over the past decade, a remarkable surge in the development of functional nano-delivery systems loaded with bioactive compounds for healthcare has been witnessed. Notably, the demanding requirements of high solubility, prolonged circulation, high tissue penetration capability, and strong targeting ability of nanocarriers have posed interdisciplinary research challenges to the community. While extensive experimental studies have been conducted to understand the construction of nano-delivery systems and their metabolic behavior in vivo, less is known about these molecular mechanisms and kinetic pathways during their metabolic process in vivo, and lacking effective means for high-throughput screening. Molecular dynamics (MD) simulation techniques provide a reliable tool for investigating the design of nano-delivery carriers encapsulating these functional ingredients, elucidating the synthesis, translocation, and delivery of nanocarriers. This review introduces the basic MD principles, discusses how to apply MD simulation to design nanocarriers, evaluates the ability of nanocarriers to adhere to or cross gastrointestinal mucosa, and regulates plasma proteins in vivo. Moreover, we presented the critical role of MD simulation in developing delivery systems for precise nutrition and prospects for the future. This review aims to provide insights into the implications of MD simulation techniques for designing and optimizing nano-delivery systems in the healthcare food industry.
Collapse
Affiliation(s)
- Li-Hang Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Jiang-Ning Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
4
|
Peng X, Zhang T, Wu Y, Wang X, Liu R, Jin X. mPEG-CS-modified flexible liposomes-reinforced thermosensitive sol-gel reversible hydrogels for ocular delivery of multiple drugs with enhanced synergism. Colloids Surf B Biointerfaces 2023; 231:113560. [PMID: 37812861 DOI: 10.1016/j.colsurfb.2023.113560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/02/2023] [Accepted: 09/17/2023] [Indexed: 10/11/2023]
Abstract
Non-invasive drug delivery offers a safe treatment while improving patient compliance. However, due to the particular physiological structure of the ocular, long-term retention and sustained drug release of the drug delivery system is crucial. Herein, this study aimed to design mPEG-CS-modified flexible liposomes-reinforced thermosensitive sol-gel reversible hydrogels (mPEG-CS-FL-TSG) for the delivery of astragaloside IV (AS-IV) and tetramethylpyrazine (TMP) to treat age-related macular degeneration. In vitro biological properties of mPEG-CS-FL and mPEG-CS-FL-TSG showed that they could be successfully taken up by ARPE-19 cells, and the uptake rate of mPEG-CS-FL-TSG was higher. Not only that, the release rate of mPEG-CS-FL-TSG was slower. More significantly, the results showed that the cytotoxicity of mPEG-CS-FL-TSG was lower than that of mPEG-CS-FL. In vivo result revealed that the drug delivery system could prominently enhance the ocular bioavailability of AS-IV and TMP, which is the enhanced synergism of well-permeable liposome and slow-releasing hydrogel. In summary, the mPEG-CS-FL-TSG can compensate for the short retention time and sudden release of liposome, as well as the low drug penetration of hydrogel, in order to show great promise in the non-invasive delivery of multiple drugs for the treatment of posterior ocular diseases.
Collapse
Affiliation(s)
- Xingru Peng
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tingting Zhang
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yujie Wu
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoyu Wang
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Liu
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xin Jin
- Department of Health Services, Logistics University of People's Armed Police Force, Tianjin, Tianjin 300162, China.
| |
Collapse
|
5
|
Huang Q, Zhu W, Gao X, Liu X, Zhang Z, Xing B. Nanoparticles-mediated ion channels manipulation: From their membrane interactions to bioapplications. Adv Drug Deliv Rev 2023; 195:114763. [PMID: 36841331 DOI: 10.1016/j.addr.2023.114763] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Ion channels are transmembrane proteins ubiquitously expressed in all cells that control various ions (e.g. Na+, K+, Ca2+ and Cl- etc) crossing cellular plasma membrane, which play critical roles in physiological processes including regulating signal transduction, cell proliferation as well as excitatory cell excitation and conduction. Abnormal ion channel function is usually associated with dysfunctions and many diseases, such as neurodegenerative disorders, ophthalmic diseases, pulmonary diseases and even cancers. The precise regulation of ion channels not only helps to decipher physiological and pathological processes, but also is expected to become cutting-edge means for disease treatment. Recently, nanoparticles-mediated ion channel manipulation emerges as a highly promising way to meet the increasing requirements with respect to their simple, efficient, precise, spatiotemporally controllable and non-invasive regulation in biomedicine and other research frontiers. Thanks the advantages of their unique properties, nanoparticles can not only directly block the pore sites or kinetics of ion channels through their tiny size effect, and perturb active voltage-gated ion channel by their charged surface, but they can also act as antennas to conduct or enhance external physical stimuli to achieve spatiotemporal, precise and efficient regulation of various ion channel activities (e.g. light-, mechanical-, and temperature-gated ion channels etc). So far, nanoparticles-mediated ion channel regulation has shown potential prospects in many biomedical fields at the interfaces of neuro- and cardiovascular modulation, physiological function regeneration and tumor therapy et al. Towards such important fields, in this typical review, we specifically outline the latest studies of different types of ion channels and their activities relevant to the diseases. In addition, the different types of stimulation responsive nanoparticles, their interaction modes and targeting strategies towards the plasma membrane ion channels will be systematically summarized. More importantly, the ion channel regulatory methods mediated by functional nanoparticles and their bioapplications associated with physiological modulation and therapeutic development will be discussed. Last but not least, current challenges and future perspectives in this field will be covered as well.
Collapse
Affiliation(s)
- Qiwen Huang
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Weisheng Zhu
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xiaoyin Gao
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xinping Liu
- School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Zhijun Zhang
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Bengang Xing
- School of Chemistry, Chemical Engineering & Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore.
| |
Collapse
|
6
|
Prabsangob N. Plant-based cellulose nanomaterials for food products with lowered energy uptake and improved nutritional value-a review. NFS JOURNAL 2023. [DOI: 10.1016/j.nfs.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
7
|
Zhao K, Xie Y, Lin X, Xu W. The Mucoadhesive Nanoparticle-Based Delivery System in the Development of Mucosal Vaccines. Int J Nanomedicine 2022; 17:4579-4598. [PMID: 36199476 PMCID: PMC9527817 DOI: 10.2147/ijn.s359118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mucosal tissue constitutes the largest interface between the body and the external environment, regulating the entry of pathogens, particles, and molecules. Mucosal immunization is the most effective way to trigger a protective mucosal immune response. However, the majority of the currently licensed vaccines are recommended to be administered by intramuscular injection, which has obvious shortcomings, such as high production costs, low patient compliance, and lack of mucosal immune response. Strategies for eliciting mucosal and systemic immune responses are being developed, including appropriate vaccine adjuvant, delivery system, and bacterial or viral vectors. Biodegradable mucoadhesive nanoparticles (NPs) are the most promising candidate for vaccine delivery systems due to their inherent immune adjuvant property and the ability to protect the antigen from degradation, sustain the release of loaded antigen, and increase the residence time of antigen at the administration site. The current review outlined the complex structure of mucosa, the mechanism of interaction between NPs and mucosa, factors affecting the mucoadhesion of NPs, and the application of the delivery system based on mucoadhesive NPs in the field of vaccines. Moreover, this review demonstrated that the biodegradable and mucoadhesive NP-based delivery system has the potential for mucosal administration of vaccines.
Collapse
Affiliation(s)
- Kai Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Correspondence: Kai Zhao, Tel +86 576 88660338, Email
| | - Yinzhuo Xie
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
| | - Xuezheng Lin
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China
- Xuezheng Lin, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, 318000, People’s Republic of China, Email
| | - Wei Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, People’s Republic of China
| |
Collapse
|
8
|
Zhang H, Yu J, Ma L, Zhao Y, Xu S, Shi J, Qian K, Gu M, Tan H, Xu L, Liu Y, Mu C, Xiong Y. Reversing multi-drug resistance by polymeric metformin to enhance antitumor efficacy of chemotherapy. Int J Pharm 2022; 624:121931. [PMID: 35750278 DOI: 10.1016/j.ijpharm.2022.121931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/01/2023]
Abstract
Multi-drug resistance (MDR) in breast cancer poses a great threat to chemotherapy. The expression and function of the ATP binding cassette (ABC) transporter are the major cause of MDR. Herein, a linear polyethylene glycol (PEI) conjugated with dicyandiamide, which called polymeric metformin (PolyMet), was successfully synthesized as a simple and biocompatible polymer of metformin. PolyMet showed the potential to reverse MDR by inhibiting the efflux of the substrate of ATP-binding cassette (ABC) transporter from DOX resistant MCF-7 cells (MCF-7/DOX). To test its MDR reversing effect, PolyMet was combined with DOX to treat mice carrying MCF-7/DOX xenografts. In order to decrease the toxicities of DOX and delivery PolyMet and DOX to tumor at the same time, PolyMet was complexed with poly-γ-glutamic acid-doxorubicin (PGA-DOX) electrostatically at the optimal ratio of 2:3, which were further coated with lipid membrane to form lipid/PolyMet-(PGA-DOX) nanoparticles (LPPD). The particle size of LPPD was 165.8 nm, and the zeta potential was +36.5 mV. LPPD exhibited favorable cytotoxicity and cellular uptake in MCF-7/DOX. Meanwhile, the bioluminescence imaging and immunohistochemical analysis indicated that LPPD effectively conquered DOX-associated MDR by blocking ABC transporters (ABCB1 and ABCC1) via PolyMet. Remarkably, LPPD significantly inhibited the tumor growth and lowered the systemic toxicity in a murine MCF-7/DOX tumor model. This is the first time to reveal that PolyMet can enhance the anti-tumor efficacy of DOX by dampening ABC transporters and activating the AMPK/mTOR pathway, which is a promising strategy for drug-resistant breast cancer therapy.
Collapse
Affiliation(s)
- Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiandong Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Lisha Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yue Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shujun Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ke Qian
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Mancang Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongsheng Tan
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Li Xu
- Zhejiang Provincial Hospital of TCM (Traditional Chinese Medicine), The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Yun Liu
- UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
9
|
Mahmoud AM, Nowell CJ, Feeney O, van 't Hag L, Davis TP, Kempe K. Hydrophobicity Regulates the Cellular Interaction of Cyanine5-Labeled Poly(3-hydroxypropionate)-Based Comb Polymers. Biomacromolecules 2022; 23:3560-3571. [PMID: 35921528 DOI: 10.1021/acs.biomac.2c00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An in-depth understanding of the effect of physicochemical properties of nanocarriers on their cellular uptake and fate is crucial for the development of novel delivery systems. In this study, well-defined hydrophobic carboxylated poly(3-hydroxypropionate)-based comb polymers were synthesized. Two oligo(3-hydroxypropionate) (HPn) of different degrees of polymerization (DP; 5 and 9) bearing α-vinyl end-groups were obtained by an hydrogen transfer polymerization (HTP)-liquid/liquid extraction strategy. 2-Carboxyethyl acrylate (CEA), representing the DP 1 analogue of HPn, was also included in the study. (Macro)monomers were polymerized via reversible addition-fragmentation chain-transfer (RAFT) polymerization and fully characterized by 1H NMR spectroscopy and size exclusion chromatography. All polymers were non-hemolytic and non-cytotoxic against NIH/3T3 cells. Detailed cellular association and uptake studies of Cy5-labeled polymers by flow cytometry and confocal laser scanning microscopy (CLSM) revealed that the carboxylated water-soluble PCEA, the polymer with the shortest side chain, efficiently targets mitochondria. However, increasing the side-chain DP led to a change in the intracellular fate. P(HP5) was trafficked to both mitochondria and lysosomes, while P(HP9) was exclusively found in lysosomes. Importantly, FLIM-FRET investigation of P(HP5) provided initial insight into the mitochondria subcompartment location of Cy5-labeled carboxylated polymers. Moreover, intracellular uptake mechanism studies were performed. Blocking scavenger receptors by dextran sulfate or cooling cells to 4 °C significantly affected the cell association of hydrophobic carboxylated polymers with an insignificant response to membrane-potential inhibitors. In contrast, water-soluble carboxylated polymers' cellular association was substantially inhibited in cells treated with compounds depleting the mitochondrial potential (ΔΨ). Overall, this study highlights hydrophobicity as a valuable means to tune the cellular interaction of carboxylated polymers and thus will inform the design of future drug carriers based on Cy5-modified carboxylated polymers.
Collapse
Affiliation(s)
- Ayaat M Mahmoud
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Orlagh Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Leonie van 't Hag
- Department of Chemical and Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Thomas P Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
10
|
Shi Y, Xu X, Yu H, Lin Z, Zuo H, Wu Y. Defined positive charge patterns created on DNA nanostructures determine cellular uptake efficiency. NANO LETTERS 2022; 22:5330-5338. [PMID: 35729707 DOI: 10.1021/acs.nanolett.2c01316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
We provide an effective method to create DNA nanostructures below 100 nm with defined charge patterns and explore whether the density and location of charges affect the cellular uptake efficiency of nanoparticles (NPs). To avoid spontaneous charge neutralization, the negatively charged polymer nanopatterns were first created by in situ polymerization using photoresponsive monomers on DNA origami. Subsequent irradiation generated positive charges on the immobilized polymers, achieving precise positively charged patterns on the negatively charged DNA surface. Via this method, we have discovered that the positive charges located on the edges of nanostructures facilitate more efficient cellular uptake in comparison to the central counterparts. In addition, the high-density positive charge decoration could also enhance particle penetration into 3D multicellular spheroids. This strategy paves a new way to construct elaborate charge-separated substructures on NP surfaces and holds great promise for a deeper understanding of the influence between the surface charge distribution and nano-bio interactions.
Collapse
Affiliation(s)
- Yiwei Shi
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Xuemei Xu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Huaibin Yu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Zian Lin
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Honghua Zuo
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Yuzhou Wu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| |
Collapse
|
11
|
Yin W, Xuan D, Wang H, Zhou M, Deng B, Ma F, Lu Y, Zhang J. Biodegradable Imiquimod-Loaded Mesoporous Organosilica as a Nanocarrier and Adjuvant for Enhanced and Prolonged Immunity against Foot-and-Mouth Disease Virus in Mice. ACS APPLIED BIO MATERIALS 2022; 5:3095-3106. [PMID: 35679606 DOI: 10.1021/acsabm.2c00382] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Foot-and-mouth disease (FMD), a serious, fast-spreading, and virulent disease, has led to huge economic losses to people all over the world. Vaccines are the most effective way to control FMD. However, the weak immunogenicity of inactivated FMD virus (FMDV) requires the addition of adjuvants to enhance the immune effectiveness of the vaccines. Herein, we formulated and fabricated biodegradable dendritic mesoporous tetrasulfide-doped organosilica nanoparticles SOMSN with imiquimod complex (SOMSN-IMQ) and used it as a platform for FMD vaccine delivery and as an adjuvant. SOMSN-IMQ demonstrated excellent stability for 6 months when stored in PBS, while it could be completely degraded within 42 days in SBF at room temperature. Biosafety experiments such as cell toxicity, hemolysis, and histology indicated that the as-prepared SOMSN-IMQ showed nontoxicity and good biocompatibility. Furthermore, SOMSN-IMQ exhibited a maximum adsorption capacity of 1000 μg/mg for inactivated FMDV antigens. Our results showed that SOMSN-IMQ can be effectively engulfed by RAW264.7 cells in a dose-dependent manner. After immunization, SOMSN-IMQ@FMDV can elicit persistent higher antibody levels, higher IgG2a/IgG1 ratio, and cytokine expression, which indicated that SOMSN-IMQ@FMDV triggered superior humoral and cellular immune responses. Moreover, SOMSN-IMQ could provoke maturation and activation of dendritic cells in lymph nodes (LDCs) as well as the proliferation of lymphocytes in vivo. Thus, SOMSN-IMQ could promote effective and potent immunity and provide a promising adjuvant platform for FMDV vaccination with acceptable safety.
Collapse
Affiliation(s)
- Wenzhu Yin
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious and Zoonoses, Yangzhou 225009, P. R. China
| | - Dechun Xuan
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China.,School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Haiyan Wang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China
| | - Mingxu Zhou
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China
| | - Bihua Deng
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China
| | - Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China.,School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious and Zoonoses, Yangzhou 225009, P. R. China
| | - Jinqiu Zhang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, P. R. China.,School of Pharmacy, Jiangsu University, Zhenjiang 212013, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious and Zoonoses, Yangzhou 225009, P. R. China
| |
Collapse
|
12
|
Anastasiadis SH, Chrissopoulou K, Stratakis E, Kavatzikidou P, Kaklamani G, Ranella A. How the Physicochemical Properties of Manufactured Nanomaterials Affect Their Performance in Dispersion and Their Applications in Biomedicine: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:552. [PMID: 35159897 PMCID: PMC8840392 DOI: 10.3390/nano12030552] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/21/2022]
Abstract
The growth in novel synthesis methods and in the range of possible applications has led to the development of a large variety of manufactured nanomaterials (MNMs), which can, in principle, come into close contact with humans and be dispersed in the environment. The nanomaterials interact with the surrounding environment, this being either the proteins and/or cells in a biological medium or the matrix constituent in a dispersion or composite, and an interface is formed whose properties depend on the physicochemical interactions and on colloidal forces. The development of predictive relationships between the characteristics of individual MNMs and their potential practical use critically depends on how the key parameters of MNMs, such as the size, shape, surface chemistry, surface charge, surface coating, etc., affect the behavior in a test medium. This relationship between the biophysicochemical properties of the MNMs and their practical use is defined as their functionality; understanding this relationship is very important for the safe use of these nanomaterials. In this mini review, we attempt to identify the key parameters of nanomaterials and establish a relationship between these and the main MNM functionalities, which would play an important role in the safe design of MNMs; thus, reducing the possible health and environmental risks early on in the innovation process, when the functionality of a nanomaterial and its toxicity/safety will be taken into account in an integrated way. This review aims to contribute to a decision tree strategy for the optimum design of safe nanomaterials, by going beyond the compromise between functionality and safety.
Collapse
Affiliation(s)
- Spiros H. Anastasiadis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
- Department of Chemistry, University of Crete, 700 13 Heraklion, Crete, Greece
| | - Kiriaki Chrissopoulou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
- Department of Physics, University of Crete, 700 13 Heraklion, Crete, Greece
| | - Paraskevi Kavatzikidou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
| | - Georgia Kaklamani
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
| | - Anthi Ranella
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, N. Plastira 100, 700 13 Heraklion, Crete, Greece; (K.C.); (E.S.); (P.K.); (G.K.); (A.R.)
| |
Collapse
|
13
|
Chen S, Chen Y, Fu M, Cao Q, Wang B, Chen W, Ma X. Active Nanomotors Surpass Passive Nanomedicines: Current Progress and Challenges. J Mater Chem B 2022; 10:7099-7107. [DOI: 10.1039/d2tb00556e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Artificial nanomotors show advantages over traditional nanomedicines in biomedical applications due to their active locomotion by converting various energy sources into mechanical force in situ. Currently, nanomotors have attracted wide...
Collapse
|
14
|
Kumar K, Rani V, Mishra M, Chawla R. New paradigm in combination therapy of siRNA with chemotherapeutic drugs for effective cancer therapy. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100103. [PMID: 35586474 PMCID: PMC9108887 DOI: 10.1016/j.crphar.2022.100103] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chemotherapeutics drugs play a pivotal role in the treatment of cancer. However, many issues generate by chemotherapy drugs, including unfavorable harm to healthy cells and multidrug resistance (MDR), persist and have a negative impact on therapeutic outcomes. When compared to monotherapy, combination cancer therapy has many advantages, like improving efficacy through synergistic effects and overcoming drug resistance. Combination treatment may comprise several chemotherapeutics drugs and combinations of chemotherapeutic drugs with some other therapeutic options such as surgery or radiation. Cancer treatment that utilizes co-delivery strategies with siRNA and chemotherapeutic drugs has been shown to have highly effective antitumor effects in the treatment of many cancers. However, the highly complex mechanisms of chemotherapeutic drugs-siRNA pairs during the co-delivery process have received little attention. The ideal combination of chemotherapeutic drugs with siRNA is very crucial for producing the desirable anticancer effects that would greatly enhance therapeutic efficiency. This review puts an emphasis on the logic for choosing suitable chemotherapeutic drug-siRNA combinations, which may open the way for the co-delivery of chemotherapeutic drugs and siRNA for treating cancer in the clinic. This review summarizes recent breakthrough in the area of diverse mechanism-based chemotherapeutic drugs-siRNA combinations in cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Ruchi Chawla
- Corresponding author. Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, U.P., India.
| |
Collapse
|
15
|
Souri M, Soltani M, Moradi Kashkooli F, Kiani Shahvandi M, Chiani M, Shariati FS, Mehrabi MR, Munn LL. Towards principled design of cancer nanomedicine to accelerate clinical translation. Mater Today Bio 2022; 13:100208. [PMID: 35198957 PMCID: PMC8841842 DOI: 10.1016/j.mtbio.2022.100208] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/08/2023] Open
Abstract
Nanotechnology in medical applications, especially in oncology as drug delivery systems, has recently shown promising results. However, although these advances have been promising in the pre-clinical stages, the clinical translation of this technology is challenging. To create drug delivery systems with increased treatment efficacy for clinical translation, the physicochemical characteristics of nanoparticles such as size, shape, elasticity (flexibility/rigidity), surface chemistry, and surface charge can be specified to optimize efficiency for a given application. Consequently, interdisciplinary researchers have focused on producing biocompatible materials, production technologies, or new formulations for efficient loading, and high stability. The effects of design parameters can be studied in vitro, in vivo, or using computational models, with the goal of understanding how they affect nanoparticle biophysics and their interactions with cells. The present review summarizes the advances and technologies in the production and design of cancer nanomedicines to achieve clinical translation and commercialization. We also highlight existing challenges and opportunities in the field.
Collapse
Key Words
- CFL, Cell-free layer
- CGMD, Coarse-grained molecular dynamic
- Clinical translation
- DPD, Dissipative particle dynamic
- Drug delivery
- Drug loading
- ECM, Extracellular matrix
- EPR, Permeability and retention
- IFP, Interstitial fluid pressure
- MD, Molecular dynamic
- MDR, Multidrug resistance
- MEC, Minimum effective concentration
- MMPs, Matrix metalloproteinases
- MPS, Mononuclear phagocyte system
- MTA, Multi-tadpole assemblies
- MTC, Minimum toxic concentration
- Nanomedicine
- Nanoparticle design
- RBC, Red blood cell
- TAF, Tumor-associated fibroblast
- TAM, Tumor-associated macrophage
- TIMPs, Tissue inhibitor of metalloproteinases
- TME, Tumor microenvironment
- Tumor microenvironment
Collapse
Affiliation(s)
- Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| | | | | | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Lance L. Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
16
|
Alallam B, Doolaanea AA, Kyaw Oo M, Mohd Nasir MH, Taher M. Influence of nanoparticles surface coating on physicochemical properties for CRISPR gene delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
17
|
Gonzalez-Valdivieso J, Garcia-Sampedro A, Hall AR, Girotti A, Arias FJ, Pereira SP, Acedo P. Smart Nanoparticles as Advanced Anti-Akt Kinase Delivery Systems for Pancreatic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55790-55805. [PMID: 34788541 DOI: 10.1021/acsami.1c14592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pancreatic cancer is one of the deadliest cancers partly due to late diagnosis, poor drug delivery to the target site, and acquired resistance to therapy. Therefore, more effective therapies are urgently needed to improve the outcome of patients. In this work, we have tested self-assembling genetically engineered polymeric nanoparticles formed by elastin-like recombinamers (ELRs), carrying a small peptide inhibitor of the protein kinase Akt, in both PANC-1 and patient-derived pancreatic cancer cells (PDX models). Nanoparticle cell uptake was measured by flow cytometry, and subcellular localization was determined by confocal microscopy, which showed a lysosomal localization of these nanoparticles. Furthermore, metabolic activity and cell viability were significantly reduced after incubation with nanoparticles carrying the Akt inhibitor in a time- and dose-dependent fashion. Self-assembling 73 ± 3.2 nm size nanoparticles inhibited phosphorylation and consequent activation of Akt protein, blocked the NF-κB signaling pathway, and triggered caspase 3-mediated apoptosis. Furthermore, in vivo assays showed that ELR-based nanoparticles were suitable devices for drug delivery purposes with long circulating time and minimum toxicity. Hence, the use of these smart nanoparticles could lead to the development of more effective treatment options for pancreatic cancer based on the inhibition of Akt.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Andres Garcia-Sampedro
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Andrew R Hall
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, United Kingdom
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| |
Collapse
|
18
|
Sohrabi Kashani A, Packirisamy M. Cancer-Nano-Interaction: From Cellular Uptake to Mechanobiological Responses. Int J Mol Sci 2021; 22:9587. [PMID: 34502495 PMCID: PMC8431109 DOI: 10.3390/ijms22179587] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
With the advancement of nanotechnology, the nano-bio-interaction field has emerged. It is essential to enhance our understanding of nano-bio-interaction in different aspects to design nanomedicines and improve their efficacy for therapeutic and diagnostic applications. Many researchers have extensively studied the toxicological responses of cancer cells to nano-bio-interaction, while their mechanobiological responses have been less investigated. The mechanobiological properties of cells such as elasticity and adhesion play vital roles in cellular functions and cancer progression. Many studies have noticed the impacts of cellular uptake on the structural organization of cells and, in return, the mechanobiology of human cells. Mechanobiological changes induced by the interactions of nanomaterials and cells could alter cellular functions and influence cancer progression. Hence, in addition to biological responses, the possible mechanobiological responses of treated cells should be monitored as a standard methodology to evaluate the efficiency of nanomedicines. Studying the cancer-nano-interaction in the context of cell mechanics takes our knowledge one step closer to designing safe and intelligent nanomedicines. In this review, we briefly discuss how the characteristic properties of nanoparticles influence cellular uptake. Then, we provide insight into the mechanobiological responses that may occur during the nano-bio-interactions, and finally, the important measurement techniques for the mechanobiological characterizations of cells are summarized and compared. Understanding the unknown mechanobiological responses to nano-bio-interaction will help with developing the application of nanoparticles to modulate cell mechanics for controlling cancer progression.
Collapse
Affiliation(s)
| | - Muthukumaran Packirisamy
- Optical Bio-Microsystem Lab, Micro-Nano-Bio-Integration Centre, Department of Mechanical, Industrial and Aerospace Engineering, Concordia University, 1455 De Maisonneuve Blvd. W., Montreal, QC H3G 1M8, Canada;
| |
Collapse
|
19
|
Radaic A, Joo NE, Jeong SH, Yoo SII, Kotov N, Kapila YL. Phosphatidylserine-Gold Nanoparticles (PS-AuNP) Induce Prostate and Breast Cancer Cell Apoptosis. Pharmaceutics 2021; 13:1094. [PMID: 34371784 PMCID: PMC8309069 DOI: 10.3390/pharmaceutics13071094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/01/2023] Open
Abstract
Prostate and breast cancer are the current leading causes of new cancer cases in males and females, respectively. Phosphatidylserine (PS) is an essential lipid that mediates macrophage efferocytosis and is dysregulated in tumors. Therefore, developing therapies that selectively restore PS may be a potential therapeutic approach for carcinogenesis. Among the nanomedicine strategies for delivering PS, biocompatible gold nanoparticles (AuNPs) have an extensive track record in biomedical applications. In this study, we synthesized biomimetic phosphatidylserine-caped gold nanoparticles (PS-AuNPs) and tested their anticancer potential in breast and prostate cancer cells in vitro. We found that both cell lines exhibited changes in cell morphology indicative of apoptosis. After evaluating for histone-associated DNA fragments, a hallmark of apoptosis, we found significant increases in DNA fragmentation upon PS-AuNP treatment compared to the control treatment. These findings demonstrate the use of phosphatidylserine coupled with gold nanoparticles as a potential treatment for prostate and breast cancer. To the best of our knowledge, this is the first time that a phosphatidylserine-capped AuNP has been examined for its therapeutic potential in cancer therapy.
Collapse
Affiliation(s)
- Allan Radaic
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (A.R.); (N.E.J.)
| | - Nam E. Joo
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (A.R.); (N.E.J.)
| | - Soo-Hwan Jeong
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea;
| | - Seong-II Yoo
- Department of Polymer Engineering, Pukyong National University, Busan 608737, Korea;
| | - Nicholas Kotov
- Department of Chemical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Yvonne L. Kapila
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (A.R.); (N.E.J.)
| |
Collapse
|
20
|
Chiarpotti MV, Longo GS, Del Pópolo MG. Nanoparticles modified with cell penetrating peptides: Assessing adsorption on membranes containing acidic lipids. Colloids Surf B Biointerfaces 2021; 197:111373. [DOI: 10.1016/j.colsurfb.2020.111373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 01/12/2023]
|
21
|
Ackun-Farmmer MA, Alatise KL, Cross G, Benoit DSW. Ligand Density Controls C-Type Lectin-Like Molecule-1 Receptor-Specific Uptake of Polymer Nanoparticles. ADVANCED BIOSYSTEMS 2020; 4:e2000172. [PMID: 33073549 PMCID: PMC7959326 DOI: 10.1002/adbi.202000172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/01/2020] [Indexed: 01/13/2023]
Abstract
The newest generation of drug delivery systems (DDSs) exploits ligands to mediate specific targeting of cells and/or tissues. However, studies investigating the link between ligand density and nanoparticle (NP) uptake are limited to a small number of ligand-receptor systems. C-type lectin-like molecule-1 (CLL1) is uniquely expressed on myeloid cells, which enables the development of receptors specifically targeting treat various diseases. This study aims to investigate how NPs with different CLL1 targeting peptide density impact cellular uptake. To this end, poly(styrene-alt-maleic anhydride)-b-poly(styrene) NPs are functionalized with cyclized CLL1 binding peptides (cCBP) ranging from 240 ± 12 to 31 000 ± 940 peptides per NP. Unexpectedly, the percentage of cells with internalized NPs is decreased for all cCBP-NP designs regardless of ligand density compared to unmodified NPs. Internalization through CLL1 receptor-mediated processes is further investigated without confounding the effects of NP size and surface charge. Interestingly, high density cCBP-NPs (>7000 cCBP per NP) uptake is dominated by CLL1 receptor-mediated processes while low density cCBP-NPs (≈200 cCBP per NP) and untargeted NP occurred through non-specific clathrin and caveolin-mediated endocytosis. Altogether, these studies show that ligand density and uptake mechanism should be carefully investigated for specific ligand-receptor systems for the design of targeted DDSs to achieve effective drug delivery.
Collapse
Affiliation(s)
- Marian A Ackun-Farmmer
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
- University of Rochester Medical Center, Department of Orthopaedics and Center for Musculoskeletal Research, Rochester, NY, USA
| | - Kharimat L Alatise
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
| | - Griffin Cross
- Washington University in St. Louis, Biomedical/Medical Engineering, St. Louis, MO, USA
| | - Danielle S W Benoit
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
- University of Rochester Medical Center, Department of Orthopaedics and Center for Musculoskeletal Research, Rochester, NY, USA
- University of Rochester, Materials Science Program, Rochester, NY, USA
- University of Rochester, Department of Chemical Engineering, Rochester, NY, USA
| |
Collapse
|
22
|
Valente KP, Suleman A, Brolo AG. Exploring Diffusion and Cellular Uptake: Charged Gold Nanoparticles in an in Vitro Breast Cancer Model. ACS APPLIED BIO MATERIALS 2020; 3:6992-7002. [PMID: 35019358 DOI: 10.1021/acsabm.0c00872] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gold nanoparticles have emerged as a prominent tool in nanomedicine, particularly for applications in cancer diagnostic and treatment. One of the challenges for the successful implementation of gold nanoparticles in cancer therapy is their delivery to the specific cancer area within the tumor microenvironment. The presence of cancer enables a poorly organized vascularization system, increasing the pressure with the microenvironment, limiting the uptake of particles. The physicochemical properties of the gold nanoparticles (size, shape, and surface charge) also have a significant effect on diffusion to the tumor site and cellular uptake. In this work, we analyzed the transport of 10 nm gold nanoparticles with different surface charges (neutral, negative, and positive) through a hydrogel composite. Three-dimensional in vitro models composed of breast cancer cells loaded in the hydrogel composite were used for the qualitative and quantitative evaluation of cellular uptake of the gold nanoparticles. Surprisingly, an inverse correlation between the diffusion coefficients of the nanoparticles and cellular uptake was demonstrated. Positively charged gold nanoparticles displayed high cellular uptake, although their diffusion coefficient indicated slow transport through the hydrogel matrix. Neutral particles, on the other hand, displayed fast diffusion but the lowest cellular uptake. The results obtained indicate that nanoparticle diffusion and cellular uptake should be studied together in realistic in vitro models for a true evaluation of transport in tumor microenvironments.
Collapse
Affiliation(s)
- Karolina P Valente
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technology, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
| | - Afzal Suleman
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
| | - Alexandre G Brolo
- Department of Chemistry, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technology, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
23
|
Hameed M, Panicker S, Abdallah SH, Khan AA, Han C, Chehimi MM, Mohamed AA. Protein-Coated Aryl Modified Gold Nanoparticles for Cellular Uptake Study by Osteosarcoma Cancer Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:11765-11775. [PMID: 32931295 DOI: 10.1021/acs.langmuir.0c01443] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Gold nanoparticles coated with proteins have shown extraordinary biocompatibility which advanced to several nanomedicine engineering applications. We synthesized protein-coated gold nanoparticles using green and chemical reduction routes for cellular uptake study. In the current work, we coated gold-aryl nanoparticles of the type AuNPs-C6H4-4-COOH with bovine serum albumin (BSA), collagen, zein, and lysozyme proteins. Both routes were carried out without phase-transfer catalysts or extraneous stabilizing agents. High crystallinity of the AuNPs synthesized by the green route can be seen in transmission electron microscopy images. Osteosarcoma cancer cells are malignant bone tumors with abnormal cellular functions. Studies using MG-63 cells will provide mechanistic suggestions on the details of the amplification in tumors. We studied the cellular uptake of the bioconjugates by MG-63 osteosarcoma cells using laser confocal fluorescence microscopy (LCFM) and flow cytometry. In the LCFM study, BSA-AuNPs were uptaken most efficiently of all protein-coated gold nanoparticles synthesized by the green route. Lysozyme-AuNPs synthesized by the chemical reduction method were mostly efficiently internalized by MG-63 cells among all AuNPs. Zein- and lysozyme-coated AuNPs, though of relatively small size, prepared by the green method were not efficiently uptaken by MG-63. The two nanoparticles are negatively charged, and zein is also a hydrophobic coat. The difference in hydrophobicity and charge might have affected the internalization. All of those coated nanoparticles that were efficiently uptaken can potentially be used as diagnostic and therapeutic agents for osteosarcoma.
Collapse
Affiliation(s)
- Mehavesh Hameed
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Seema Panicker
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sallam H Abdallah
- Human Genetics and Stem Cells Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amir A Khan
- Human Genetics and Stem Cells Research Group, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Applied Biology, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Changseok Han
- Department of Environmental Engineering, INHA University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Mohamed M Chehimi
- Université Paris-Est Créteil, CNRS, ICMPE, UMR7182, F-94320 Thiais, France
| | - Ahmed A Mohamed
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
24
|
Genova J, Chamati H, Petrov M. Study of SOPC with embedded pristine and amide-functionalized single wall carbon nanotubes by DSC and FTIR spectroscopy. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.125261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
25
|
Fatima N, Gromnicova R, Loughlin J, Sharrack B, Male D. Gold nanocarriers for transport of oligonucleotides across brain endothelial cells. PLoS One 2020; 15:e0236611. [PMID: 32941446 PMCID: PMC7498062 DOI: 10.1371/journal.pone.0236611] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/31/2020] [Indexed: 12/04/2022] Open
Abstract
Treatment of diseases that affect the CNS by gene therapy requires delivery of oligonucleotides to target cells within the brain. As the blood brain barrier prevents movement of large biomolecules, current approaches involve direct injection of the oligonucleotides, which is invasive and may have only a localised effect. The aim of this study was to investigate the potential of 2 nm galactose-coated gold nanoparticles (NP-Gal) as a delivery system of oligonucleotides across brain endothelium. DNA oligonucleotides of different types were attached to NP-Gal by the place exchange reaction and were characterised by EMSA (electrophoretic mobility shift assay). Several nanoparticle formulations were created, with single- or double-stranded (20nt or 40nt) DNA oligonucleotides, or with different amounts of DNA attached to the carriers. These nanocarriers were applied to transwell cultures of human brain endothelium in vitro (hCMEC/D3 cell-line) or to a 3D-hydrogel model of the blood-brain barrier including astrocytes. Transfer rates were measured by quantitative electron microscopy for the nanoparticles and qPCR for DNA. Despite the increase in nanoparticle size caused by attachment of oligonucleotides to the NP-Gal carrier, the rates of endocytosis and transcytosis of nanoparticles were both considerably increased when they carried an oligonucleotide cargo. Carriers with 40nt dsDNA were most efficient, accumulating in vesicles, in the cytosol and beneath the basal membrane of the endothelium. The oligonucleotide cargo remained attached to the nanocarriers during transcytosis and the transport rate across the endothelial cells was increased at least 50fold compared with free DNA. The nanoparticles entered the extracellular matrix and were taken up by the astrocytes in biologically functional amounts. Attachment of DNA confers a strong negative charge to the nanoparticles which may explain the enhanced binding to the endothelium and transcytosis by both vesicular transport and the transmembrane/cytosol pathway. These gold nanoparticles have the potential to transport therapeutic amounts of nucleic acids into the CNS.
Collapse
Affiliation(s)
- Nayab Fatima
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Radka Gromnicova
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Jane Loughlin
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Basil Sharrack
- Academic Department of Neuroscience and Sheffield, NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals, NHS Foundation Trust, University of Sheffield, Sheffield, United Kingdom
| | - David Male
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
26
|
Rueda-Gensini L, Cifuentes J, Castellanos MC, Puentes PR, Serna JA, Muñoz-Camargo C, Cruz JC. Tailoring Iron Oxide Nanoparticles for Efficient Cellular Internalization and Endosomal Escape. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1816. [PMID: 32932957 PMCID: PMC7559083 DOI: 10.3390/nano10091816] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Iron oxide nanoparticles (IONs) have been widely explored for biomedical applications due to their high biocompatibility, surface-coating versatility, and superparamagnetic properties. Upon exposure to an external magnetic field, IONs can be precisely directed to a region of interest and serve as exceptional delivery vehicles and cellular markers. However, the design of nanocarriers that achieve an efficient endocytic uptake, escape lysosomal degradation, and perform precise intracellular functions is still a challenge for their application in translational medicine. This review highlights several aspects that mediate the activation of the endosomal pathways, as well as the different properties that govern endosomal escape and nuclear transfection of magnetic IONs. In particular, we review a variety of ION surface modification alternatives that have emerged for facilitating their endocytic uptake and their timely escape from endosomes, with special emphasis on how these can be manipulated for the rational design of cell-penetrating vehicles. Moreover, additional modifications for enhancing nuclear transfection are also included in the design of therapeutic vehicles that must overcome this barrier. Understanding these mechanisms opens new perspectives in the strategic development of vehicles for cell tracking, cell imaging and the targeted intracellular delivery of drugs and gene therapy sequences and vectors.
Collapse
Affiliation(s)
- Laura Rueda-Gensini
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Javier Cifuentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Maria Claudia Castellanos
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Julian A. Serna
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
27
|
Jiao F, Sang J, Liu Z, Liu W, Liang W. Effect of concentration of PEG coated gold nanoparticle on lung surfactant studied with coarse-grained molecular dynamics simulations. Biophys Chem 2020; 266:106457. [PMID: 32890945 DOI: 10.1016/j.bpc.2020.106457] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 11/25/2022]
Abstract
The surface modification of nanoparticles can not only change the physical and chemical properties of particles, such as the hydrophilic and hydrophobic properties and surface charges of nanoparticles to a certain extent, but also bring new functions to nanoparticles, such as membrane permeability and targeting. Inhaled nanoparticles (NPs) are experienced by the first biological barrier inside the alveolus known as lung surfactant (LS), consisting of phospholipids and proteins in the form of the monolayer at the air-water interface. Inhaled NPs can reach deep into the lungs and interfere with the biophysical properties of the lung components. The interaction mechanisms of bare gold nanoparticles (AuNPs) with the LS monolayer are not well understood. Coarse-grained molecular dynamics simulations were carried out to have a study on the interactions of PEG coated AuNPs with LS monolayers. It was observed that the interactions of AuNPs and LS components make the monolayer structure deform and change the biophysical properties of LS monolayer. The results also indicate that AuNPs with high concentrations hinder the lowering of the LS surface tension and reduce lateral mobility of lipids. Overall, the simulation results can provide guidance for the design of ligand protected NPs as drug carriers and can identify the nanoparticles potential side effect on lung surfactant.
Collapse
Affiliation(s)
- Fengxuan Jiao
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, PR China
| | - Jianbing Sang
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, PR China.
| | - Zhaoyang Liu
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, PR China
| | - Wei Liu
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, PR China.
| | - Weiguang Liang
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, PR China
| |
Collapse
|
28
|
Real time monitoring of interactions of gold nanoparticles with supported phospholipid lipid layers. J Electroanal Chem (Lausanne) 2020. [DOI: 10.1016/j.jelechem.2020.114302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
29
|
Zartner L, Garni M, Craciun I, Einfalt T, Palivan CG. How Can Giant Plasma Membrane Vesicles Serve as a Cellular Model for Controlled Transfer of Nanoparticles? Biomacromolecules 2020; 22:106-115. [PMID: 32648740 DOI: 10.1021/acs.biomac.0c00624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cellular model systems are essential platforms used across multiple research fields for exploring the fundaments of biology and biochemistry. Here, we present giant plasma membrane vesicles (GPMVs) as a platform of cell-like compartments that will facilitate the study of particles within a biorelevant environment and promote their further development. We studied how cellularly taken up nanoparticles (NPs) can be transferred into formed GPMVs and which are the molecular factors that play a role in successful transfer (size, concentration, and surface charge along with 3 different cell lines: HepG2, HeLa, and Caco-2). We observed that polystyrene (PS) carboxylated NPs with a size of 40 and 100 nm were successfully and efficiently transferred to GPMVs derived from all cell lines. We then investigated the distribution of NPs inside formed GPMVs and established the average number of NPs/GPMVs and the percentage of all GPMVs with NPs in their cavity. We pave the way for GPMV usage as superior cell-like mimics in medically relevant applications.
Collapse
Affiliation(s)
- Luisa Zartner
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, P.O. Box 3350, CH-4002 Basel, Switzerland
| | - Martina Garni
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, P.O. Box 3350, CH-4002 Basel, Switzerland
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, P.O. Box 3350, CH-4002 Basel, Switzerland
| | - Tomaž Einfalt
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, P.O. Box 3350, CH-4002 Basel, Switzerland
| |
Collapse
|
30
|
Engstrom AM, Faase RA, Marquart GW, Baio JE, Mackiewicz MR, Harper SL. Size-Dependent Interactions of Lipid-Coated Gold Nanoparticles: Developing a Better Mechanistic Understanding Through Model Cell Membranes and in vivo Toxicity. Int J Nanomedicine 2020; 15:4091-4104. [PMID: 32606666 PMCID: PMC7295544 DOI: 10.2147/ijn.s249622] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/13/2020] [Indexed: 01/13/2023] Open
Abstract
Introduction Humans are intentionally exposed to gold nanoparticles (AuNPs) where they are used in variety of biomedical applications as imaging and drug delivery agents as well as diagnostic and therapeutic agents currently in clinic and in a variety of upcoming clinical trials. Consequently, it is critical that we gain a better understanding of how physiochemical properties such as size, shape, and surface chemistry drive cellular uptake and AuNP toxicity in vivo. Understanding and being able to manipulate these physiochemical properties will allow for the production of safer and more efficacious use of AuNPs in biomedical applications. Methods and Materials Here, AuNPs of three sizes, 5 nm, 10 nm, and 20 nm, were coated with a lipid bilayer composed of sodium oleate, hydrogenated phosphatidylcholine, and hexanethiol. To understand how the physical features of AuNPs influence uptake through cellular membranes, sum frequency generation (SFG) was utilized to assess the interactions of the AuNPs with a biomimetic lipid monolayer composed of a deuterated phospholipid 1.2-dipalmitoyl-d62-sn-glycero-3-phosphocholine (dDPPC). Results and Discussion SFG measurements showed that 5 nm and 10 nm AuNPs are able to phase into the lipid monolayer with very little energetic cost, whereas, the 20 nm AuNPs warped the membrane conforming it to the curvature of hybrid lipid-coated AuNPs. Toxicity of the AuNPs were assessed in vivo to determine how AuNP curvature and uptake influence cell health. In contrast, in vivo toxicity tested in embryonic zebrafish showed rapid toxicity of the 5 nm AuNPs, with significant 24 hpf mortality occurring at concentrations ≥20 mg/L, whereas the 10 nm and 20 nm AuNPs showed no significant mortality throughout the five-day experiment. Conclusion By combining information from membrane models using SFG spectroscopy with in vivo toxicity studies, a better mechanistic understanding of how nanoparticles (NPs) interact with membranes is developed to understand how the physiochemical features of AuNPs drive nanoparticle-membrane interactions, cellular uptake, and toxicity.
Collapse
Affiliation(s)
- Arek M Engstrom
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Ryan A Faase
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR, United States
| | - Grant W Marquart
- Department of Chemistry, Portland State University, Portland, OR, United States
| | - Joe E Baio
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR, United States
| | | | - Stacey L Harper
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States.,School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR, United States.,Oregon Nanoscience and Microtechnologies Institute, Corvallis, OR, United States
| |
Collapse
|
31
|
Villanueva-Flores F, Castro-Lugo A, Ramírez OT, Palomares LA. Understanding cellular interactions with nanomaterials: towards a rational design of medical nanodevices. NANOTECHNOLOGY 2020; 31:132002. [PMID: 31770746 PMCID: PMC7105107 DOI: 10.1088/1361-6528/ab5bc8] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 05/05/2023]
Abstract
Biomedical applications increasingly require fully characterized new nanomaterials. There is strong evidence showing that nanomaterials not only interact with cells passively but also actively, mediating essential molecular processes for the regulation of cellular functions, but we are only starting to understand the mechanisms of those interactions. Systematic studies about cell behavior as a response to specific nanoparticle properties are scarce in the literature even when they are necessary for the rational design of medical nanodevices. Information in the literature shows that the physicochemical properties determine the bioactivity, biocompatibility, and safety of nanomaterials. The information available regarding the interaction and responses of cells to nanomaterials has not been analyzed and discussed in a single document. Hence, in this review, we present the latest advances about cellular responses to nanomaterials and integrate the available information into concrete considerations for the development of innovative, efficient, specific and, more importantly, safe biomedical nanodevices. We focus on how physicochemical nanoparticle properties (size, chemical surface, shape, charge, and topography) influence cell behavior in a first attempt to provide a practical guide for designing medical nanodevices, avoiding common experimental omissions that may lead to data misinterpretation. Finally, we emphasize the importance of the systematic study of nano-bio interactions to acquire sufficient reproducible information that allows accurate control of cell behavior based on tuning of nanomaterial properties. This information is useful to guide the design of specific nanodevices and nanomaterials to elicit desired cell responses, like targeting, drug delivery, cell attachment, differentiation, etc, or to avoid undesired side effects.
Collapse
Affiliation(s)
- Francisca Villanueva-Flores
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Andrés Castro-Lugo
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Octavio T Ramírez
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| | - Laura A Palomares
- Instituto de Biotecnología. Universidad Nacional Autónoma de México. Ave. Universidad 2001. Col. Chamilpa. Cuernavaca, Morelos 62210, México
Villanueva-Flores F: ; Castro-Lugo A: ; Ramírez O: ; Palomares L:
| |
Collapse
|
32
|
Kumar Basak U, Roobala C, Basu JK, Maiti PK. Size-dependent interaction of hydrophilic/hydrophobic ligand functionalized cationic and anionic nanoparticles with lipid bilayers. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2020; 32:104003. [PMID: 31722322 DOI: 10.1088/1361-648x/ab5770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We study the nature of nanoparticle (NPs)-membrane interaction as a function of nanoparticle size for different functionalization using molecular dynamics simulation. Zinc sulphide quantum dots of size, 2 nm and 4 nm are used as model NPs, and DLPC and DPPC lipid bilayers are used as model membranes. We use coarse-grained polarizable MARTINI model (MPW) to simulate the NPs and lipid bilayers. Our simulation results show that uncharged bare NPs penetrate the lipid bilayers and embed themselves within the hydrophobic core of the bilayer both in the gel and fluid phases. NPs of size 4 nm are shown to disrupt the bilayer. The bilayer recovers from the damages caused by smaller NPs of size 2 nm. In case of either purely hydrophilic or hybrid (with hydrophilic/hydrophobic ratio of 2:1) ligand-functionalized NPs of smaller size (shell size 2 nm), only cationic NPs bind to the bilayer. However, for larger NPs with a shell size of 4 nm, both anionic and cationic hybrid functionalized NPs bind to the bilayer. The performance of standard Martini (SM) force field for the charged NP/bilayer systems has also been tested and compared with the results obtained using MPW model. Although the overall trend that the cationic NPs interact strongly with the bilayers than their anionic counterparts has been captured correctly using SM, the adsorption behaviour of the functionalized NPs differ significantly in the SM force field. The interaction of anionic NPs with both fluid and gel bilayers has been observed to be least accurately represented in the SM force field.
Collapse
|
33
|
Bai X, Li M, Hu G. Nanoparticle translocation across the lung surfactant film regulated by grafting polymers. NANOSCALE 2020; 12:3931-3940. [PMID: 32003385 DOI: 10.1039/c9nr09251j] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nanoparticle-based pulmonary drug delivery has gained significant attention due to its ease of administration, increased bioavailability, and reduced side effects caused by a high systemic dosage. After being delivered into the deep lung, the inhaled nanoparticles first interact with the lung surfactant lining layer composed of phospholipids and surfactant proteins and then potentially cause the dysfunction of the lung surfactant. Conditioning the surface properties of nanoparticles with grafting polymers to avoid these side effects is of crucial importance to the efficiency and safety of pulmonary drug delivery. Herein, we perform coarse-grained molecular simulations to decipher the involved mechanism responsible for the translocation of the polymer-grafted Au nanoparticles across the lung surfactant film. The simulations illustrate that conditioning of the grafting polymers, including their length, terminal charge, and grafting density, can result in different translocation processes. Based on the energy analysis, we find that these discrepancies in translocation stem from the affinity of the nanoparticles with the lipid tails and heads and their contact with the proteins, which can be tuned by the surface polarity and surface charge of the nanoparticles. We further demonstrate that the interaction between the nanoparticles and the lung surfactant is related to the depletion of the lipids and proteins during translocation, which affects the surface tension of the surfactant film. The change in the surface tension in turn affects the nanoparticle translocation and the collapse of the surfactant film. These results can help understand the adverse effects of the nanoparticles on the lung surfactant film and provide guidance to the design of inhaled nanomedicines for improved permeability and targeting.
Collapse
Affiliation(s)
- Xuan Bai
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China. and The State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mujun Li
- The State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China and School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoqing Hu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
34
|
Uskoković V. X-ray photoelectron and ion scattering spectroscopic surface analyses of amorphous and crystalline calcium phosphate nanoparticles with different chemical histories. Phys Chem Chem Phys 2020; 22:5531-5547. [PMID: 32123882 DOI: 10.1039/c9cp06529f] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The surface of hydroxyapatite nanoparticles is enriched in the topmost atomic layer with calcium and depleted of it elsewhere, alongside being dependent on the history of formation of hydroxyapatite from the amorphous precursor.
Collapse
Affiliation(s)
- Vuk Uskoković
- Department of Mechanical and Aerospace Engineering
- University of California Irvine
- Irvine
- USA
| |
Collapse
|
35
|
Zuraw-Weston S, Wood DA, Torres IK, Lee Y, Wang LS, Jiang Z, Lázaro GR, Wang S, Rodal AA, Hagan MF, Rotello VM, Dinsmore AD. Nanoparticles binding to lipid membranes: from vesicle-based gels to vesicle tubulation and destruction. NANOSCALE 2019; 11:18464-18474. [PMID: 31577313 PMCID: PMC7155749 DOI: 10.1039/c9nr06570a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
While cells offer numerous inspiring examples in which membrane morphology and function are controlled by interactions with viruses or proteins, we still lack design principles for controlling membrane morphology in synthetic systems. With experiments and simulations, we show that spherical nanoparticles binding to lipid-bilayer membrane vesicles results in a remarkably rich set of collective morphologies that are controllable via the particle binding energy. We separately study cationic and anionic particles, where the adhesion is tuned by addition of oppositely charged lipids to the vesicles. When the binding energy is weak relative to a characteristic membrane-bending energy, vesicles adhere to one another and form a soft solid gel, a novel and useful platform for controlled release. With larger binding energy, a transition from partial to complete wrapping of the nanoparticles causes a remarkable vesicle destruction process culminating in rupture, nanoparticle-membrane tubules, and an apparent inversion of the vesicles. These findings help unify the diverse phenomena observed previously. They also open the door to a new class of vesicle-based, closed-cell gels that are more than 99% water and can encapsulate and release on demand, and show how to drive intentional membrane remodeling for shape-responsive systems.
Collapse
|
36
|
Wang Y, Han X, Cui Z, Shi D. Bioelectricity, Its Fundamentals, Characterization Methodology, and Applications in Nano-Bioprobing and Cancer Diagnosis. ACTA ACUST UNITED AC 2019; 3:e1900101. [PMID: 32648718 DOI: 10.1002/adbi.201900101] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/01/2019] [Indexed: 12/11/2022]
Abstract
Bioelectricity is an essential characteristic of a biological system that has played an important role in medical diagnosis particularly in cancer liquid biopsy. However, its biophysical origin and measurements have presented great challenges in experimental methodologies. For instance, in dynamic cell processes, bioelectricity cannot be accurately determined as a static electrical potential via electrophoresis. Cancer cells fundamentally differ from normal cells by having a much higher rate of glycolysis resulting in net negative charges on cell surfaces. The most recent investigations on cancer cell surface charge that is the direct bio-electrical manifestation of the "Warburg Effect," which can be directly monitored by specially designed nanoprobes, has been provided. The most up-to-date research results from charge-mediated cell targeting are reviewed. Correlations between the cell surface charge and cancer cell metabolism are established based on cell/probe electrostatic interactions. Bioelectricity is utilized not only as an analyte for investigation of the metabolic state of the cancer cells, but also applied in electrostatically and magnetically capturing of the circulating tumor cells from whole blood. Also reviewed is on the isolation of Candida albicans via bioelectricity-driven nanoparticle binding on fungus with surface charges.
Collapse
Affiliation(s)
- Yilong Wang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| | - Xiao Han
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China
| | - Zheng Cui
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, P. R. China.,Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Donglu Shi
- Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| |
Collapse
|
37
|
Shimokawa N, Ito H, Higuchi Y. Coarse-grained molecular dynamics simulation for uptake of nanoparticles into a charged lipid vesicle dominated by electrostatic interactions. Phys Rev E 2019; 100:012407. [PMID: 31499808 DOI: 10.1103/physreve.100.012407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Indexed: 06/10/2023]
Abstract
We use a coarse-grained molecular dynamics simulation to investigate the interaction between neutral or charged nanoparticles (NPs) and a vesicle consisting of neutral and negatively charged lipids. We focus on the interaction strengths of hydrophilic and hydrophobic attraction and electrostatic interactions between a lipid molecule and an NP. A neutral NP passes through the lipid membrane when the hydrophobic interaction is sufficiently strong. As the valence of the positively charged NP increases, the membrane permeation speed of the NP is increased compared with the neutral NP and charged lipids are accumulated around the charged NP. A charged NP with a high valence passes through the lipid membrane via a transient channel formed by charged lipids or transportlike endocytosis. These permeation processes can be classified based on analyses of the density correlation function. When the nonelectrostatic interaction parameters are large enough, a negatively charged NP can be adsorbed on the membrane and a neutral lipid-rich region is formed directly below the NP. The NP is spontaneously incorporated into the vesicle under various conditions and the incorporation is mediated by the membrane curvature. We reveal how the NP's behavior depends on the NP valence, size, and the nonelectrostatic interaction parameters.
Collapse
Affiliation(s)
- Naofumi Shimokawa
- School of Materials Science, Japan Advanced Institute of Science and Technology, Ishikawa 923-1292, Japan
| | - Hiroaki Ito
- Department of Mechanical Engineering, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Yuji Higuchi
- Institute for Solid State Physics, University of Tokyo, Chiba 227-8581, Japan
| |
Collapse
|
38
|
Gonzalez-Valdivieso J, Girotti A, Muñoz R, Rodriguez-Cabello JC, Arias FJ. Self-Assembling ELR-Based Nanoparticles as Smart Drug-Delivery Systems Modulating Cellular Growth via Akt. Biomacromolecules 2019; 20:1996-2007. [DOI: 10.1021/acs.biomac.9b00206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Juan Gonzalez-Valdivieso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Raquel Muñoz
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - J. Carlos Rodriguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - F. Javier Arias
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| |
Collapse
|
39
|
Wang S, Guo H, Li Y, Li X. Penetration of nanoparticles across a lipid bilayer: effects of particle stiffness and surface hydrophobicity. NANOSCALE 2019; 11:4025-4034. [PMID: 30768108 DOI: 10.1039/c8nr09381d] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The cellular uptake of nanoparticles (NPs) has drawn significant attention due to their great importance and potential in drug delivery, bioimaging, and specific targeting. Here, we conduct a computational study on the translocation process of soft nanoparticles with different elasticities and surface hydrophobicities through a lipid bilayer membrane. It is shown that the translocation abilities of hydrophilic NPs can be enhanced by increasing their stiffness, while the penetrability of hydrophobic NPs is weakened by increasing the particle stiffness. The free energy analysis indicates that rigid hydrophilic NPs and soft hydrophobic NPs encounter lower energy barriers during penetration. In direct translocation, different deformation modes are observed for NPs with different surface hydrophobicities during cellular internalization. Further, deformation analysis demonstrates that hydrophilic NPs are flattened in the membrane plane, while hydrophobic NPs are elongated along the membrane norm during penetration. We conclude that the elasticity of NPs has an obvious impact on their ability to penetrate across the lipid bilayer membrane through different morphological responses of hydrophilic and hydrophobic NPs. These results shed light on the coupled effects of particle elasticity and surface hydrophobicity on the cellular uptake of elastic NPs, which may provide useful guidelines for designing effective nanocarrier systems for drug delivery.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering (State Key Laboratory of Ocean Engineering, MOE Key Laboratory of Hydrodynamics), Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Hui Guo
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yinfeng Li
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering (State Key Laboratory of Ocean Engineering, MOE Key Laboratory of Hydrodynamics), Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Xuejin Li
- Department of Engineering Mechanics and Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou 310027, P. R. China.
| |
Collapse
|
40
|
Lázaro GR, Mukhopadhyay S, Hagan MF. Why Enveloped Viruses Need Cores-The Contribution of a Nucleocapsid Core to Viral Budding. Biophys J 2019; 114:619-630. [PMID: 29414708 DOI: 10.1016/j.bpj.2017.11.3782] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/11/2017] [Accepted: 11/27/2017] [Indexed: 11/17/2022] Open
Abstract
During the lifecycle of many enveloped viruses, a nucleocapsid core buds through the cell membrane to acquire an outer envelope of lipid membrane and viral glycoproteins. However, the presence of a nucleocapsid core is not required for assembly of infectious particles. To determine the role of the nucleocapsid core, we develop a coarse-grained computational model with which we investigate budding dynamics as a function of glycoprotein and nucleocapsid interactions, as well as budding in the absence of a nucleocapsid. We find that there is a transition between glycoprotein-directed budding and nucleocapsid-directed budding that occurs above a threshold strength of nucleocapsid interactions. The simulations predict that glycoprotein-directed budding leads to significantly increased size polydispersity and particle polymorphism. This polydispersity can be explained by a theoretical model accounting for the competition between bending energy of the membrane and the glycoprotein shell. The simulations also show that the geometry of a budding particle leads to a barrier to subunit diffusion, which can result in a stalled, partially budded state. We present a phase diagram for this and other morphologies of budded particles. Comparison of these structures against experiments could establish bounds on whether budding is directed by glycoprotein or nucleocapsid interactions. Although our model is motivated by alphaviruses, we discuss implications of our results for other enveloped viruses.
Collapse
Affiliation(s)
- Guillermo R Lázaro
- Martin Fisher School of Physics, Brandeis University, Waltham, Massachusetts
| | | | - Michael F Hagan
- Martin Fisher School of Physics, Brandeis University, Waltham, Massachusetts.
| |
Collapse
|
41
|
Zolghadr AR, Moosavi SS. Interactions of neutral gold nanoparticles with DPPC and POPC lipid bilayers: simulation and experiment. RSC Adv 2019; 9:5197-5205. [PMID: 35514645 PMCID: PMC9060696 DOI: 10.1039/c8ra06777e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/21/2019] [Indexed: 11/21/2022] Open
Abstract
Molecular dynamics simulations of neutral gold nanoparticles (AuNPs) interacting with dipalmitoylphosphatidylcholine (DPPC) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) membranes were studied using a model system.
Collapse
|
42
|
Raman AS, Pajak J, Chiew Y. Interaction of PCL based self-assembled nano-polymeric micelles with model lipid bilayers using coarse-grained molecular dynamics simulations. Chem Phys Lett 2018. [DOI: 10.1016/j.cplett.2018.09.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Foroozandeh P, Aziz AA. Insight into Cellular Uptake and Intracellular Trafficking of Nanoparticles. NANOSCALE RESEARCH LETTERS 2018; 13:339. [PMID: 30361809 PMCID: PMC6202307 DOI: 10.1186/s11671-018-2728-6] [Citation(s) in RCA: 762] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/24/2018] [Indexed: 05/06/2023]
Abstract
Nanoparticle science is rapidly changing the landscape of various scientific fields and defining new technological platforms. This is perhaps even more evident in the field of nanomedicine whereby nanoparticles have been used as a tool for the treatment and diagnosis of many diseases. However, despite the tremendous benefit conferred, common pitfalls of this technology is its potential short and long-term effects on the human body. To understand these issues, many scientific studies have been carried out. This review attempts to shed light on some of these studies and its outcomes. The topics that were examined in this review include the different possible uptake pathways of nanoparticles and intracellular trafficking routes. Additionally, the effect of physicochemical properties of nanoparticle such as size, shape, charge and surface chemistry in determining the mechanism of uptake and biological function of nanoparticles are also addressed.
Collapse
Affiliation(s)
- Parisa Foroozandeh
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang Malaysia
| | - Azlan Abdul Aziz
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang Malaysia
- Nano-Biotechnology Research and Innovation (NanoBRI), Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Gelugor, Penang Malaysia
| |
Collapse
|
44
|
Ojha PK, Kar S, Roy K, Leszczynski J. Toward comprehension of multiple human cells uptake of engineered nano metal oxides: quantitative inter cell line uptake specificity (QICLUS) modeling. Nanotoxicology 2018; 13:14-34. [PMID: 30354872 DOI: 10.1080/17435390.2018.1529836] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To address the nanomaterial exposure threat, it is imperative to understand how nanomaterials are recognized, internalized, and distributed within diverse cell systems. Targeting of nanomaterials to a specific cell type is generally attained through the modification of the nanoparticle (NP) surface leading to required cellular uptake. The enhanced cellular uptake to normal cells can direct to the higher interaction of NPs with subcellular organelles resulting the provocation of various signaling pathways. The successes of NPs rely on the prospect for the synthesis of functionalized NPs with necessary properties and their enhanced potential for cellular uptake for specific targeting. In the present study, we have modeled the cellular uptake of 109 surface modifiers of metal oxide nanoparticles (MNPs) for three different cell lines: HUVEC (Human endothelial cells), U937 (human macrophage cells), and PaCa2 (cancer cell lines). Along with the quantitative structure-activity relationship (QSAR) models, for the very first time we have developed and performed quantitative inter cell line uptake specificity (QICLUS) modeling to identify the physicochemical properties, as well as majorly structural fragments responsible for cellular uptake differences between two specific cell lines. The present work provides a comprehensive understanding of the cellular uptake of MNPs and the underlying structural parameters controlling the nano-cellular interactions. This phenomenon has also been analyzed from the QSAR and QICLUS models that concluded the functional groups of surface modifiers like amine, anhydride, halogen atoms, nitro group, acids have the dominating roles for the uptake of MNPs into the cell lines. Thus, the developed models may be used for designing of novel surface modifiers of MNPs of desired characteristics for proper cell-NPs interactions, as well as in the context of virtual screening aspect. Moreover, the MNP-cell interactions can give some idea about the toxicity for target-specific drug delivery treatment as higher cellular uptake is required for specific cells to treat the disease and lower uptake to the neighboring cells for lower toxicity.
Collapse
Affiliation(s)
- Probir Kumar Ojha
- a Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - Supratik Kar
- b Interdisciplinary Nanotoxicity Center, Department of Chemistry, Physics and Atmospheric Sciences , Jackson State University , Jackson , MS , USA
| | - Kunal Roy
- a Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - Jerzy Leszczynski
- b Interdisciplinary Nanotoxicity Center, Department of Chemistry, Physics and Atmospheric Sciences , Jackson State University , Jackson , MS , USA
| |
Collapse
|
45
|
Shi X, Tian F. Multiscale Modeling and Simulation of Nano‐Carriers Delivery through Biological Barriers—A Review. ADVANCED THEORY AND SIMULATIONS 2018. [DOI: 10.1002/adts.201800105] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xinghua Shi
- CAS Key Laboratory for Nanosystem and Hierarchy FabricationCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyChinese Academy of Sciences Beijing 100190 China
- School of Nanoscience and TechnologyUniversity of Chinese Academy of Sciences NO.19A Yuquan Road Beijing 100049 China
| | - Falin Tian
- CAS Key Laboratory for Nanosystem and Hierarchy FabricationCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyChinese Academy of Sciences Beijing 100190 China
- School of Nanoscience and TechnologyUniversity of Chinese Academy of Sciences NO.19A Yuquan Road Beijing 100049 China
| |
Collapse
|
46
|
Urbančič I, Garvas M, Kokot B, Majaron H, Umek P, Cassidy H, Škarabot M, Schneider F, Galiani S, Arsov Z, Koklic T, Matallanas D, Čeh M, Muševič I, Eggeling C, Štrancar J. Nanoparticles Can Wrap Epithelial Cell Membranes and Relocate Them Across the Epithelial Cell Layer. NANO LETTERS 2018; 18:5294-5305. [PMID: 30039976 PMCID: PMC6089500 DOI: 10.1021/acs.nanolett.8b02291] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/24/2018] [Indexed: 06/08/2023]
Abstract
Although the link between the inhalation of nanoparticles and cardiovascular disease is well established, the causal pathway between nanoparticle exposure and increased activity of blood coagulation factors remains unexplained. To initiate coagulation tissue factor bearing epithelial cell membranes should be exposed to blood, on the other side of the less than a micrometre thin air-blood barrier. For the inhaled nanoparticles to promote coagulation, they need to bind lung epithelial-cell membrane parts and relocate them into the blood. To assess this hypothesis, we use advanced microscopy and spectroscopy techniques to show that the nanoparticles wrap themselves with epithelial-cell membranes, leading to the membrane's disruption. The membrane-wrapped nanoparticles are then observed to freely diffuse across the damaged epithelial cell layer relocating epithelial cell membrane parts over the epithelial layer. Proteomic analysis of the protein content in the nanoparticles wraps/corona finally reveals the presence of the coagulation-initiating factors, supporting the proposed causal link between the inhalation of nanoparticles and cardiovascular disease.
Collapse
Affiliation(s)
- Iztok Urbančič
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Weatherall
Institute of Molecular Medicine, University
of Oxford, Headley Way, Oxford OX3
9DS, United Kingdom
| | - Maja Garvas
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Jožef
Stefan International Postgraduate School, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Boštjan Kokot
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Hana Majaron
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Polona Umek
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Center
of Excellence NAMASTE, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Hilary Cassidy
- Systems
Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Miha Škarabot
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Falk Schneider
- Weatherall
Institute of Molecular Medicine, University
of Oxford, Headley Way, Oxford OX3
9DS, United Kingdom
| | - Silvia Galiani
- Weatherall
Institute of Molecular Medicine, University
of Oxford, Headley Way, Oxford OX3
9DS, United Kingdom
| | - Zoran Arsov
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Center
of Excellence NAMASTE, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Tilen Koklic
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Center
of Excellence NAMASTE, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - David Matallanas
- Systems
Biology Ireland, University College Dublin, Dublin 4, Ireland
- School of
Medicine and Medical Science, University
College Dublin, Dublin 4, Ireland
| | - Miran Čeh
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Igor Muševič
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Faculty
of Mathematics and Physics, University of
Ljubljana, Jadranska
19, SI-1000 Ljubljana, Slovenia
| | - Christian Eggeling
- Weatherall
Institute of Molecular Medicine, University
of Oxford, Headley Way, Oxford OX3
9DS, United Kingdom
- Institute
of Applied Optics, Friedrich-Schiller University, Jena 07749, Germany
- Leibniz
Institute of Photonic Technology (IPHT), Jena 07745, Germany
| | - Janez Štrancar
- “Jožef
Stefan Institute”, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
- Center
of Excellence NAMASTE, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
47
|
Zhao J, Stenzel MH. Entry of nanoparticles into cells: the importance of nanoparticle properties. Polym Chem 2018. [DOI: 10.1039/c7py01603d] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Knowledge of the interactions between nanoparticles (NPs) and cell membranes is of great importance for the design of safe and efficient nanomedicines.
Collapse
Affiliation(s)
- Jiacheng Zhao
- Centre for Advanced Macromolecular Design
- The University of New South Wales
- Sydney
- Australia
- School of Chemical Engineering
| | - Martina H. Stenzel
- Centre for Advanced Macromolecular Design
- The University of New South Wales
- Sydney
- Australia
- School of Chemistry
| |
Collapse
|
48
|
Quan X, Zhao D, Li L, Zhou J. Understanding the Cellular Uptake of pH-Responsive Zwitterionic Gold Nanoparticles: A Computer Simulation Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:14480-14489. [PMID: 29166558 DOI: 10.1021/acs.langmuir.7b03544] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Surface functionalization of nanoparticles (NPs) with stealth polymers (e.g., hydrophilic and zwitterionic polymers) has become a common strategy to resist nonspecific protein adsorption recently. Understanding the role of surface decoration on NP-biomembrane interactions is of great significance to promote the application of NPs in biomedical fields. Herein, using coarse-grained molecular dynamics (CGMD) simulations, we investigate the interactions between stealth polymer-coated gold nanoparticles (AuNPs) and lipid membranes. The results show that AuNPs grafted with zwitterionic polymers can more easily approach the membrane surface than those coated with hydrophilic poly(ethylene glycol) (PEG), which can be explained by the weak dipole-dipole interaction between them. For zwitterionic AuNPs which can undergo pH-dependent charge conversion, different interaction modes which depend on the polymer protonation degree are found. When the protonation degree is low, the particles just adsorb on the membrane surface; at moderate protonation degrees, the particles can directly translocate across the lipid membrane through a transient hydrophilic pore formed on the membrane surface; the particles are fully wrapped by the curved lipid membrane at high protonation degrees, which may lead to endocytosis. Finally, the effect of polymer chain length on the cellular uptake of zwitterionic polymer-coated AuNPs is considered. The results demonstrate that longer polymer chain length will block the translocation of AuNPs across the lipid membrane when the protonation degree is not high; however, it can improve the transmembrane efficiency of AuNPs at high protonation degrees. We expect that these findings are of immediate interest to the design and synthesis of pH-responsive nanomaterials based on zwitterionic polymers and can prompt their further applications in the field of biomedicine.
Collapse
Affiliation(s)
- Xuebo Quan
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab for Green Chemical Product Technology, South China University of Technology , Guangzhou 510640, P. R. China
| | - Daohui Zhao
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab for Green Chemical Product Technology, South China University of Technology , Guangzhou 510640, P. R. China
| | - Libo Li
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab for Green Chemical Product Technology, South China University of Technology , Guangzhou 510640, P. R. China
| | - Jian Zhou
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Lab for Green Chemical Product Technology, South China University of Technology , Guangzhou 510640, P. R. China
| |
Collapse
|
49
|
Zhang Z, Lin X, Gu N. Effects of temperature and PEG grafting density on the translocation of PEGylated nanoparticles across asymmetric lipid membrane. Colloids Surf B Biointerfaces 2017; 160:92-100. [PMID: 28918189 DOI: 10.1016/j.colsurfb.2017.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/20/2017] [Accepted: 09/05/2017] [Indexed: 01/19/2023]
Abstract
Plasma membrane internalization of nanoparticles (NPs) is important for their biomedical applications such as drug-delivery carriers. On one hand, in order to improve their half-life in circulation, PEGylation has been widely used. However, it may hinder the NPs' membrane internalization ability. On the other hand, higher temperature could enhance the membrane permeability and may affect the NPs' ability to enter into or exit from cells. To make full use of their advantages, we systematically investigated the effects of temperature and PEG density on the translocation of PEGylated nanoparticles across the plasma asymmetric membrane of eukaryotic cells, using near-atom level coarse-grained molecular dynamics simulations. Our results showed that higher temperature could accelerate the translocation of NPs across membranes by making lipids more disorder and faster diffusion. On the contrary, steric hindrance effects of PEG would inhibit NPs' translocation process and promote lipids flip-flops. The PEG chains could rearrange themselves to minimize the contacts between PEG and lipid tails during the translocation, which was similar to 'snorkeling effect'. Moreover, lipid flip-flops were affected by PEGylated density as well as NPs' translocation direction. Higher PEG grafting density could promote lipid flip-flops, but inhibit lipid extraction from bilayers. The consequence of lipid flip-flop and extraction was that the membranes got more symmetric.
Collapse
Affiliation(s)
- Zuoheng Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Bio materials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano-Science and Technology, Suzhou Key Laboratory of Biomaterials and Technologies, Suzhou 215123, PR China
| | - Xubo Lin
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Bio materials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Department of Integrative Biology & Pharmacology, Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Bio materials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano-Science and Technology, Suzhou Key Laboratory of Biomaterials and Technologies, Suzhou 215123, PR China.
| |
Collapse
|
50
|
Behzadi S, Serpooshan V, Tao W, Hamaly MA, Alkawareek MY, Dreaden EC, Brown D, Alkilany AM, Farokhzad OC, Mahmoudi M. Cellular uptake of nanoparticles: journey inside the cell. Chem Soc Rev 2017; 46:4218-4244. [PMID: 28585944 PMCID: PMC5593313 DOI: 10.1039/c6cs00636a] [Citation(s) in RCA: 1466] [Impact Index Per Article: 209.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoscale materials are increasingly found in consumer goods, electronics, and pharmaceuticals. While these particles interact with the body in myriad ways, their beneficial and/or deleterious effects ultimately arise from interactions at the cellular and subcellular level. Nanoparticles (NPs) can modulate cell fate, induce or prevent mutations, initiate cell-cell communication, and modulate cell structure in a manner dictated largely by phenomena at the nano-bio interface. Recent advances in chemical synthesis have yielded new nanoscale materials with precisely defined biochemical features, and emerging analytical techniques have shed light on nuanced and context-dependent nano-bio interactions within cells. In this review, we provide an objective and comprehensive account of our current understanding of the cellular uptake of NPs and the underlying parameters controlling the nano-cellular interactions, along with the available analytical techniques to follow and track these processes.
Collapse
Affiliation(s)
- Shahed Behzadi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|