201
|
Zhen X, Cheng P, Pu K. Recent Advances in Cell Membrane-Camouflaged Nanoparticles for Cancer Phototherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1804105. [PMID: 30457701 DOI: 10.1002/smll.201804105] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/03/2018] [Indexed: 05/28/2023]
Abstract
Phototherapy including photothermal therapy (PTT) and photodynamic therapy (PDT) employs phototherapeutic agents to generate heat or cytotoxic reactive oxygen species (ROS), and has therefore garnered particular interest for cancer therapy. However, the main challenges faced by conventional phototherapeutic agents include easy recognition by the immune system, rapid clearance from blood circulation, and low accumulation in target sites. Cell-membrane coating has emerged as a potential way to overcome these limitations, owing to the abundant proteins on the surface of cell membranes that can be inherited to the cell membrane-camouflaged nanoparticles. This review summarizes the recent advances in the development of biomimetic cell membrane-camouflaged nanoparticles for cancer phototherapy. Different sources of cell membranes can be used to coat nanoparticles uisng different coating approaches. After cell-membrane coating, the photophysical properties of the original phototherapeutic nanoparticles remain nearly unchanged; however, the coated nanoparticles are equipped with additional physiological features including immune escape, in vivo prolonged circulation time, or homologous targeting, depending on the cell sources. Moreover, the coated cell membrane can be ablated from phototherapeutic nanoparticles under laser irradiation, leading to drug release and thus synergetic therapy. By combining other supplementary agents to normalize tumor microenvironment, cell-membrane coating can further enhance the therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Xu Zhen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Penghui Cheng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| |
Collapse
|
202
|
Extracellular vesicles for personalized medicine: The input of physically triggered production, loading and theranostic properties. Adv Drug Deliv Rev 2019; 138:247-258. [PMID: 30553953 DOI: 10.1016/j.addr.2018.12.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022]
Abstract
Emerging advances in extracellular vesicle (EV) research brings along new promises for tailoring clinical treatments in order to meet specific disease features of each patient in a personalized medicine concept. EVs may act as regenerative effectors conveying endogenous therapeutic factors from parent cells or constitute a bio-camouflaged delivery system for exogenous therapeutic agents. Physical stimulation may be an important tool in the field of EVs for personalized therapy by powering EV production, loading and therapeutic properties. Physically-triggered EV production is inspired by naturally occurring EV release by shear stress in blood vessels. Bioinspired physically-triggered EV production technologies may bring along high yield advantages combined to scalability assets. Physical stimulation may also provide new prospects for high-efficient EV loading. Additionally, physically-triggered EV theranostic properties brings new hopes for spatio-temporal controlled therapy combined to tracking. Technological considerations related to EV-based personalized medicine and the input of physical stimulation on EV production, loading and theranostic properties will be overviewed herein.
Collapse
|
203
|
Kroll AV, Jiang Y, Zhou J, Holay M, Fang RH, Zhang L. Biomimetic Nanoparticle Vaccines for Cancer Therapy. ADVANCED BIOSYSTEMS 2019; 3:e1800219. [PMID: 31728404 PMCID: PMC6855307 DOI: 10.1002/adbi.201800219] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Indexed: 12/25/2022]
Abstract
It is currently understood that, in order for a tumor to successfully grow, it must evolve means of evading immune surveillance. In the past several decades, researchers have leveraged increases in our knowledge of tumor immunology to develop therapies capable of augmenting endogenous immunity and eliciting strong antitumor responses. In particular, the goal of anticancer vaccination is to train the immune system to properly utilize its own resources in the fight against cancer. Although attractive in principle, there are currently only limited examples of anticancer vaccines that have been successfully translated to the clinic. Recently, there has been a significant push towards the use of nanotechnology for designing vaccine candidates that exhibit enhanced potency and specificity. In this progress report, we discuss recent developments in the field of anticancer nanovaccines. By taking advantage of the flexibility offered by nanomedicine to purposefully program immune responses, this new generation of vaccines has the potential to address many of the hurdles facing traditional platforms. A specific emphasis is placed on the emergence of cell membrane-coated nanoparticles, a novel biomimetic platform that can be used to generate personalized nanovaccines that elicit strong, multi-antigenic antitumor responses.
Collapse
Affiliation(s)
- Ashley V Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jiarong Zhou
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
204
|
Deng G, Sun Z, Li S, Peng X, Li W, Zhou L, Ma Y, Gong P, Cai L. Cell-Membrane Immunotherapy Based on Natural Killer Cell Membrane Coated Nanoparticles for the Effective Inhibition of Primary and Abscopal Tumor Growth. ACS NANO 2018; 12:12096-12108. [PMID: 30444351 DOI: 10.1021/acsnano.8b05292] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Developing effective immunotherapies with low toxicity and high tumor specificity is the ultimate goal in the battle against cancer. Here, we reported a cell-membrane immunotherapy strategy that was able to eliminate primary tumors and inhibited distant tumors by using natural killer (NK) cell membrane cloaked photosensitizer 4,4',4'',4'''-(porphine-5,10,15,20-tetrayl) tetrakis (benzoic acid) (TCPP)-loaded nanoparticles (NK-NPs). The proteomic profiling of NK cell membranes was performed through shotgun proteomics, and we found that NK cell membranes enabled the NK-NPs to target tumors and could induce or enhance pro-inflammatory M1-macrophages polarization to produce antitumor immunity. The TCPP loaded in NK-NPs could induce cancer cell death through photodynamic therapy and consequently enhanced the antitumor immunity efficiency of the NK cell membranes. The results confirmed that NK-NPs selectively accumulated in the tumor and were able to eliminate primary tumor growth and produce an abscopal effect to inhibit distant tumors. This cell-membrane immunotherapeutic approach offers a strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Zhihong Sun
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Sanpeng Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xinghua Peng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Wenjun Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Lihua Zhou
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Lab for Health Informatics , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| |
Collapse
|
205
|
Xu X, Yang G, Xue X, Lu H, Wu H, Huang Y, Jing D, Xiao W, Tian J, Yao W, Pan CX, Lin TY, Li Y. A polymer-free, biomimicry drug self-delivery system fabricated via a synergistic combination of bottom-up and top-down approaches. J Mater Chem B 2018; 6:7842-7853. [PMID: 31380107 PMCID: PMC6676892 DOI: 10.1039/c8tb01464g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Compared to conventional carrier-assistant drug delivery systems (DDSs), drug self-delivery systems (DSDSs) have advantages of unprecedented drug loading capacity, minimized carrier-related toxicity and ease of preparation. However, the colloidal stability and blood circulation time of DSDSs still need to be improved. Here we report on the development of a novel biomimicry drug self-delivery system by the integration of a top-down cell membrane complexing technique into our self-delivery multifunctional nano-platform made from bottom-up approach that contains 100% active pharmaceutical ingredients (API) of Pheophorbide A and Irinotecan conjugates (named PI). Compared to conventional cell membrane coated nanoparticles with polymer framework as core and relatively low drug loading, this system consisting of red blood cell membrane vesicles complexed PI (RBC-PI) is polymer-free with up to 50% API loading. RBC-PI exhibited 10 times higher area under curve in pharmacokinetic study and much lower macrophage uptake compared with the parent PI nanoparticles. RBC-PI retained the excellent chemophototherapeutic effects of the PI nanoparticles, but possessed superior anti-cancer efficacy with prolonged blood circulation, improved tumor delivery, and enhanced photothermal effects in animal models. This system represents a novel example of using cell membrane complexing technique for effective surface modification of DSDSs. This is also an innovative study to form a polymer-free cell membrane nanoparticle complexing with positive surface charged materials. This biomimicry DSDS takes advantages of the best features from both systems to make up for each other's shortcomings and posed all the critical features for an ideal drug delivery system.
Collapse
Affiliation(s)
- Xiaobao Xu
- College of Biomedical Engineering & Instrument Science,
Zhejiang University, Hangzhou 310027, China
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Gaomai Yang
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Hongwei Lu
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Yee Huang
- Institute of Animal Husbandry and Veterinary Science,
Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, China
| | - Di Jing
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Jingkui Tian
- College of Biomedical Engineering & Instrument Science,
Zhejiang University, Hangzhou 310027, China
| | - Wei Yao
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Chong-xian Pan
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Tzu-yin Lin
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| |
Collapse
|
206
|
Xu L, Wu S, Wang J. Cancer cell membrane–coated nanocarriers for homologous target inhibiting the growth of hepatocellular carcinoma. J BIOACT COMPAT POL 2018. [DOI: 10.1177/0883911518819107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Lei Xu
- Beihua University, Jilin, China
| | - Shuo Wu
- Beihua University, Jilin, China
| | - Jun Wang
- Department of Hepatobiliary Surgery, The Central Hospital of Jilin City, Jilin, China
| |
Collapse
|
207
|
Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release 2018; 294:102-113. [PMID: 30553849 DOI: 10.1016/j.jconrel.2018.12.019] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Owing to the diversity and ease of preparation of nanomaterials, the rational nanocarriers with a rational design have become increasingly popular in medical researches. Although nanoparticle-based drug delivery exhibits great potential, there are some challenges facing like rapid plasma clearance, triggering or aggravation of immune response, etc. Herein, cell-based targeted drug delivery systems have drawn more and more attention owing to low immunogenicity and intrinsic mutation rate, and innate ability to allow targeted delivery. Mesenchymal stem cells (MSCs) have been used in gene and drug delivery. The use of MSCs is a promising approach for the development of gene transfer systems and drug loading strategies because of their intrinsic properties, including homing ability and tumor tropism. By combining the inherent cell properties and merits of synthetic nanoparticles (NPs), cell membrane coated NPs emerge as the time requires. Overall, we provide a comprehensive overview of the utility of MSCs in drug and gene delivery as well as MSC membrane coated nanoparticles for therapy and drug delivery, aiming to figure out the significant room for development and highlight the potential future directions.
Collapse
|
208
|
Wang YY, Wang WL, Shen XC, Zhou B, Chen T, Guo ZX, Wen CC, Jiang BP, Liang H. Combination-Responsive MoO 3- x-Hybridized Hyaluronic Acid Hollow Nanospheres for Cancer Phototheranostics. ACS APPLIED MATERIALS & INTERFACES 2018; 10:42088-42101. [PMID: 30408413 DOI: 10.1021/acsami.8b15818] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
It is of extreme importance to reduce side effects resulting from the nonspecific uptake of phototherapeutic agents by normal tissues. Currently, the single responsive strategy still cannot entirely satisfy the requirements of practical applications. In this study, we developed one kind of combination-responsive phototherapeutic nanoplatforms, where oxygen-deficient molybdenum oxide (MoO3- x) hybridized hyaluronic acid (HA) hollow nanospheres, namely, MoO3- x@HA HNSs, were constructed via a facile one-step method. In MoO3- x@HA HNSs, the reasonable combination of actively targeted specificity endowed by the HA component and tumor microenvironment-responsive phototherapy activity induced by the MoO3- x component can effectively improve the precision of phototherapy. The in vitro and in vivo experimental results confirm that MoO3- x@HA HNSs can selectively kill CD44-overexpressing cancer cells and inhibit tumor growth under an 808 nm laser irradiation, revealing their remarkable synergistic photothermal therapy/photodynamic therapy effect with CD44 receptor-targeted specificity and pH responsiveness in treating cancer. We also prove that MoO3- x@HA HNSs can serve as one kind of contrast agent to achieve the computed tomography/photoacoustic imaging. Encouraged by these results, it is anticipated that the reasonable combination of active targeting and tumor microenvironment responsiveness can be a promising strategy to develop phototherapeutic nanoplatforms for precise multimodality cancer theranostics.
Collapse
Affiliation(s)
- Yuan-Yuan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Wen-Long Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Bo Zhou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Ting Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Zheng-Xi Guo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Chang-Chun Wen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Bang-Ping Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science , Guangxi Normal University , Guilin 541004 , P. R. China
| |
Collapse
|
209
|
Zhuang J, Ying M, Spiekermann K, Holay M, Zhang Y, Chen F, Gong H, Lee JH, Gao W, Fang RH, Zhang L. Biomimetic Nanoemulsions for Oxygen Delivery In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1804693. [PMID: 30294884 PMCID: PMC6487258 DOI: 10.1002/adma.201804693] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/02/2018] [Indexed: 05/18/2023]
Abstract
Blood transfusion is oftentimes required for patients suffering from acute trauma or undergoing surgical procedures in order to help maintain the body's oxygen levels. The continued demand worldwide for blood products is expected to put significant strain on available resources and infrastructure. Unfortunately, efforts to develop viable alternatives to human red blood cells for transfusion are generally unsuccessful. Here, a hybrid natural-synthetic nanodelivery platform that combines the biocompatibility of the natural RBC membrane with the oxygen-carrying ability of perfluorocarbons is reported. The resulting formulation can be stored long-term and exhibits a high capacity for oxygen delivery, helping to mitigate the effects of hypoxia in vitro. In an animal model of hemorrhagic shock, mice are resuscitated at an efficacy comparable to whole blood infusion. By leveraging the advantageous properties of its constituent parts, this biomimetic oxygen delivery system may have the potential to address a critical need in the clinic.
Collapse
Affiliation(s)
- Jia Zhuang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Man Ying
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kevin Spiekermann
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yue Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Fang Chen
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hua Gong
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joo Hee Lee
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
210
|
Yue J, Wang Z, Shao D, Chang Z, Hu R, Li L, Luo SZ, Dong WF. Cancer cell membrane-modified biodegradable mesoporous silica nanocarriers for berberine therapy of liver cancer. RSC Adv 2018; 8:40288-40297. [PMID: 35558223 PMCID: PMC9091357 DOI: 10.1039/c8ra07574c] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/11/2018] [Indexed: 01/10/2023] Open
Abstract
Berberine (Ber) is regarded as a new, active and natural anti-cancer product; however, its clinical application has been limited due to its low aqueous solubility, poor gastrointestinal absorption, short residence time and poor targeting abilities. Hence, we reported a biomimetic nanoparticle as a drug delivery system to surmount these obstacles. We fabricated disulfide (S-S)-bridged mesoporous organosilica nanoparticles (ss-MONs) for Ber loading, which possessed uniform morphology, controllable mesoporous properties, highly-efficient drug loading capacity and superior biocompatibility. More interestingly, ss-MONs exhibited effective biodegradability under glutathione conditions through the breakage of the disulfide bond in ss-MONs, which promoted the Ber release. After coating human liver cancer HepG2 cell membranes (CM) on the surface of ss-MONs, the obtained CM-ss-MONs-Ber enhanced accumulation in liver cancer tissue through homologous targeting and effectively avoiding rapid blood clearance. Our findings indicate that CM-ss-MONs might be desirable drug carriers to promote the clinical use of Ber against liver cancer.
Collapse
Affiliation(s)
- Juan Yue
- College of Chemistry and Materials Science, The Key Laboratory of Functional Molecular Solids, Ministry of Education, Anhui Laboratory of Molecular-Based Materials, Center for Nano Science and Technology, Anhui Normal University No. 1, Beijing East Road Wuhu 241000 PR China
| | - Zheng Wang
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| | - Dan Shao
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| | - Zhimin Chang
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| | - Rui Hu
- Department of Radiation Oncology, Affiliated Suzhou Hospital of Nanjing Medical University China
| | - Li Li
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| | - Shi-Zhong Luo
- College of Chemistry and Materials Science, The Key Laboratory of Functional Molecular Solids, Ministry of Education, Anhui Laboratory of Molecular-Based Materials, Center for Nano Science and Technology, Anhui Normal University No. 1, Beijing East Road Wuhu 241000 PR China
| | - Wen-Fei Dong
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| |
Collapse
|
211
|
Bose RJC, Uday Kumar S, Zeng Y, Afjei R, Robinson E, Lau K, Bermudez A, Habte F, Pitteri SJ, Sinclair R, Willmann JK, Massoud TF, Gambhir SS, Paulmurugan R. Tumor Cell-Derived Extracellular Vesicle-Coated Nanocarriers: An Efficient Theranostic Platform for the Cancer-Specific Delivery of Anti-miR-21 and Imaging Agents. ACS NANO 2018; 12:10817-10832. [PMID: 30346694 PMCID: PMC6684278 DOI: 10.1021/acsnano.8b02587] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
MicroRNAs are critical regulators of cancer initiation, progression, and dissemination. Extensive evidence suggests that the inhibition of over-expressed oncogenic miRNA function can be a robust strategy for anticancer therapy. However, in vivo targeted delivery of miRNA therapeutics to various types of cancers remains a major challenge. Inspired by their natural synthesis and cargo delivery capabilities, researchers have exploited tumor cell-derived extracellular vesicles (TEVs) for the cancer-targeted delivery of therapeutics and theranostics. Here, we investigate a TEV-based nanoplatform for multimodal miRNA delivery and phototherapy treatments as well as the magnetic resonance imaging of cancer. We demonstrated loading of anti-miR-21 that blocks the function of endogenous oncogenic miR-21 over-expressed in cancer cells into and subsequent delivery by TEVs derived from 4T1 cells. We also produced Cy5-anti-miR-21-loaded TEVs from two other cancer cell lines (HepG2 and SKBR3) and confirmed their robust homologous and heterologous transfection efficiency and intracellular Cy5-anti-miR-21 delivery. Additionally, TEV-mediated anti-miR-21 delivery attenuated doxorubicin (DOX) resistance in breast cancer cells with a 3-fold higher cell kill efficiency than in cells treated with DOX alone. We then investigated TEVs as a biomimetic source for the functionalization of gold-iron oxide nanoparticles (GIONs) and demonstrated nanotheranostic properties of TEV-GIONs in vitro. TEV-GIONs demonstrated excellent T2 contrast in in vitro magnetic resonance (MR) imaging and resulted in efficient photothermal effect in 4T1 cells. We also evaluated the biodistribution and theranostic property of anti-miR-21 loaded TEV-GIONs in vivo by labeling with indocyanine green near-infrared dye. We further validated the tumor specific accumulation of TEV-GIONs using MR imaging. Our findings demonstrate that the distribution pattern of the TEV-anti-miR-21-GIONs correlated well with the tumor-targeting capability as well as the activity and efficacy obtained in response to doxorubicin combination treatments. TEVs and TEV-GIONs are promising nanotheranostics for future applications in cancer molecular imaging and therapy.
Collapse
Affiliation(s)
| | | | - Yitian Zeng
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | | | | | | | | | | | | - Robert Sinclair
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | | | | - Sanjiv S Gambhir
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | |
Collapse
|
212
|
Ren W, Sha H, Yan J, Wu P, Yang J, Li R, Zhang H, Yu L, Qian H, Liu B. Enhancement of radiotherapeutic efficacy for esophageal cancer by paclitaxel-loaded red blood cell membrane nanoparticles modified by the recombinant protein anti-EGFR-iRGD. J Biomater Appl 2018; 33:707-724. [PMID: 30388386 DOI: 10.1177/0885328218809019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Paclitaxel is widely used as a radiosensitizer for various tumors, including esophageal cancer, but its therapeutic effect remains to be improved. In this study, we constructed a novel nano-radiosensitizer, anti-EGFR-iRGD-conjugated (iE)-PRNPs, by conjugating the recombinant protein anti-epidermal growth factor receptor (EGFR)-internalizing arginine-glycine-aspartic (iRGD) to the surface of paclitaxel-loaded red blood cell membrane nanoparticles (PRNPs). The iE-PRNPs were confirmed to possess tumor-targeting, high penetrability, and sustained release properties that free paclitaxel does not possess. Compared with that of paclitaxel, the sensitizer enhancement ratio of iE-PRNPs was significantly increased (1.32-fold and 1.25-fold) in esophageal cancer cells with high and low expression levels of EGFR, respectively. Additionally, compared with that of unmodified PRNPs, the sensitizer enhancement ratio of iE-PRNPs in EGFR-overexpressing esophageal cancer cells was significantly increased (1.27-fold), while that of PRNPs in esophageal cancer cells with a low EGFR expression level increased slightly (1.06-fold). The improved radiosensitization effect was associated with enhanced G2/M arrest, increased reactive oxygen species, and more effective induction of DNA double-strand breaks. In summary, iE-PRNPs appear to be a novel type of radiosensitizer with the potential to overcome the bottleneck of esophageal cancer radiotherapeutic efficacy.
Collapse
Affiliation(s)
- Wei Ren
- 1 The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Huizi Sha
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jing Yan
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Puyuan Wu
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ju Yang
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rutian Li
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hang Zhang
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lixia Yu
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanqing Qian
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Baorui Liu
- 1 The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
213
|
Wang C, Wang Y, Zhang L, Miron RJ, Liang J, Shi M, Mo W, Zheng S, Zhao Y, Zhang Y. Pretreated Macrophage-Membrane-Coated Gold Nanocages for Precise Drug Delivery for Treatment of Bacterial Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1804023. [PMID: 30285289 DOI: 10.1002/adma.201804023] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/21/2018] [Indexed: 05/29/2023]
Abstract
Pathogenic bacterial infections and drug resistance make it urgent to develop new antibacterial agents with targeted delivery. Here, a new targeting delivery nanosystem is designed based on the potential interaction between bacterial recognizing receptors on macrophage membranes and distinct pathogen-associated molecular patterns in bacteria. Interestingly, the expression of recognizing receptors on macrophage membranes increases significantly when cultured with specific bacteria. Therefore, by coating pretreated macrophage membrane onto the surface of a gold-silver nanocage (GSNC), the nanosystem targets bacteria more efficiently. Previously, it has been shown that GSNC alone can serve as an effective antibacterial agent owing to its photothermal effect under near-infrared (NIR) laser irradiation. Furthermore, the nanocage can be utilized as a delivery vehicle for antibacterial drugs since the gold-silver nanocage presents a hollow interior and porous wall structure. With significantly improved bacterial adherence, the Sa-M-GSNC nanosystem, developed within this study, is effectively delivered and retained at the infection site both via local or systemic injections; the system also shows greatly prolonged blood circulation time and excellent biocompatibility. The present work described here is the first to utilize bacterial pretreated macrophage membrane receptors in a nanosystem to achieve specific bacterial-targeted delivery, and provides inspiration for future therapy based on this concept.
Collapse
Affiliation(s)
- Can Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Yulan Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Lingling Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Richard J Miron
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Jianfei Liang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Miusi Shi
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Wenting Mo
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Shihang Zheng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Yanbing Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
214
|
Jiang Y, Chekuri S, Fang RH, Zhang L. Engineering biological interactions on the nanoscale. Curr Opin Biotechnol 2018; 58:1-8. [PMID: 30390535 DOI: 10.1016/j.copbio.2018.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/14/2018] [Indexed: 01/28/2023]
Abstract
Nanoparticulate platforms have contributed significantly to the field of biomedical research, demonstrating advantages over traditional modalities in areas such as drug delivery, detoxification, and vaccination. When it comes to the design of nanoparticles, biomimetic strategies have become increasingly popular as a means of promoting effective interactions with biological systems. A recently developed cell membrane-coated nanoparticle platform can leverage the natural interactions that cells engage in with other cells, the extracellular matrix, and biomolecules in order to reduce undesirable nonspecific interactions, while increasing target-specific interactions. Here, we discuss the current state of these biomimetic nanoparticles and highlight how they can be used for various biomedical applications.
Collapse
Affiliation(s)
- Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanam Chekuri
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
215
|
Zhang L, Wang Z, Zhang Y, Cao F, Dong K, Ren J, Qu X. Erythrocyte Membrane Cloaked Metal-Organic Framework Nanoparticle as Biomimetic Nanoreactor for Starvation-Activated Colon Cancer Therapy. ACS NANO 2018; 12:10201-10211. [PMID: 30265804 DOI: 10.1021/acsnano.8b05200] [Citation(s) in RCA: 274] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Shutting down glucose supply by glucose oxidase (GOx) to starve tumors has been considered to be an attractive strategy in cancerous starvation therapy. Nevertheless, the in vivo applications of GOx-based starvation therapy are severely restricted by the poor GOx delivery efficiency and the self-limiting therapeutic effect. Herein, a biomimetic nanoreactor has been fabricated for starvation-activated cancer therapy by encapsulating GOx and prodrug tirapazamine (TPZ) in an erythrocyte membrane cloaked metal-organic framework (MOF) nanoparticle (TGZ@eM). The fabricated TGZ@eM nanoreactor can assist the delivery of GOx to tumor cells and then exhaust endogenous glucose and O2 to starve tumors efficiently. Importantly, the resulting tumor hypoxia by GOx-based starvation therapy further initiates the activation of TPZ, which is released from the nanoreactor in the acid lyso/endosome environment, for enhanced colon cancer therapy. More importantly, by integrating the biomimetic surface modification, the immunity-escaping and prolonged blood circulation characteristics endow our nanoreactor dramatically improved cancer targeting ability. The in vitro and in vivo outcomes indicate our biomimetic nanoreactor exhibits a strong synergistic cascade effect for colon cancer therapy in an accurate and facile manner.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
- University of Chinese Academy of Sciences , Beijing 100039 , P.R. China
| | - Zhenzhen Wang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
- University of Chinese Academy of Sciences , Beijing 100039 , P.R. China
| | - Yan Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
- University of Chinese Academy of Sciences , Beijing 100039 , P.R. China
| | - Fangfang Cao
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
- University of Chinese Academy of Sciences , Beijing 100039 , P.R. China
| | - Kai Dong
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun 130022 , P.R. China
| |
Collapse
|
216
|
Chen H, Sha H, Zhang L, Qian H, Chen F, Ding N, Ji L, Zhu A, Xu Q, Meng F, Yu L, Zhou Y, Liu B. Lipid insertion enables targeted functionalization of paclitaxel-loaded erythrocyte membrane nanosystem by tumor-penetrating bispecific recombinant protein. Int J Nanomedicine 2018; 13:5347-5359. [PMID: 30254439 PMCID: PMC6141126 DOI: 10.2147/ijn.s165109] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background There is currently much interest in cancer cell targeting and tumor penetrating for research and therapeutic purposes. Purpose To improve targeting delivery of antitumor drugs to gastric cancer, in this study, a tumor-targeting biocompatible drug delivery system derived from erythrocyte membrane for delivering paclitaxel (PTX) was constructed. Methods Erythrocyte membrane of human red blood cells (RBCs) were used for preparing of erythrocyte membrane-derived vesicles. 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-(maleimide[polyethylene glycol]-3400) (DSPE-PEG-MAL), a phospholipid derivative, was used to insert tumor-targeting molecular into erythrocyte membrane-derived vesicles. A lipid insertion method was used to functionalize these vesicles without the need for direct chemical conjugation. Furthermore, a tumor-penetrating bispecific recombinant protein named anti-EGFR-iRGD was used for the first time in this work to enable nanosystem to target and penetrate efficiently into the tumor site. Results Paclitaxel (PTX)-loaded anti-EGFR-iRGD-modified erythrocyte membrane nano-system (anti-EGFR-iRGD-RBCm-PTX, abbreviated to PRP) were manufactured. PRP was spheroid, uniformly size, about 171.7±4.7 nm in average, could be stable in vitro for 8 days, and released PTX in a biphasic pattern. PRP showed comparable cytotoxicity toward human gastric cancer cells in vitro. In vivo studies showed that, PRP accumulated in tumor site within 2 h of administration, lasted longer than 48 h, and the tumor volume was reduced 61% by PRP treatment in Balb/c nude mice, without causing severe side effects. Conclusion PRP has potential applications in cancer treatment and as an adjunct for other anticancer strategies.
Collapse
Affiliation(s)
- Hong Chen
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People's Republic of China, .,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China, .,Department of Oncology, Yixing People's Hospital, Jiangsu, People's Republic of China,
| | - Huizi Sha
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Lianru Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Fangjun Chen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Naiqin Ding
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Liulian Ji
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Anqing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Yan Zhou
- Department of Oncology, Yixing People's Hospital, Jiangsu, People's Republic of China,
| | - Baorui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People's Republic of China, .,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| |
Collapse
|
217
|
Jiao M, Zhang P, Meng J, Li Y, Liu C, Luo X, Gao M. Recent advancements in biocompatible inorganic nanoparticles towards biomedical applications. Biomater Sci 2018; 6:726-745. [PMID: 29308496 DOI: 10.1039/c7bm01020f] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Due to their intrinsic physical properties potentially useful for imaging and therapy as well as their highly engineerable surface, biocompatible inorganic nanoparticles offer novel platforms to develop advanced diagnostic and therapeutic agents for improved detection and more efficacious treatment of major diseases. The in vivo application of inorganic nanoparticles was demonstrated more than two decades ago, however it turns out to be very complicated as nanomaterials exhibit much more sophisticated pharmacokinetic properties than conventional drugs. In this review, we first discuss the in vivo behavior of inorganic nanoparticles after systematic administration, including the basic requirements for nanoparticles to be used in vivo, the impact of the particles' physicochemical properties on their pharmacokinetics, and the effects of the protein corona formed across the nano-bio interface. Next, we summarize the state-of-the-art of the preparation of biocompatible inorganic nanoparticles and bioconjugation strategies for obtaining target-specific nanoprobes. Then, the advancements in sensitive tumor imaging towards diagnosis and visualization of the abnormal signatures in the tumor microenvironment, together with recent studies on atherosclerosis imaging are highlighted. Finally, the future challenges and the potential for inorganic nanoparticles to be translated into clinical applications are discussed.
Collapse
Affiliation(s)
- Mingxia Jiao
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China.
| | | | | | | | | | | | | |
Collapse
|
218
|
Rudnitzki F, Feineis S, Rahmanzadeh R, Endl E, Lutz J, Groll J, Hüttmann G. siRNA release from gold nanoparticles by nanosecond pulsed laser irradiation and analysis of the involved temperature increase. JOURNAL OF BIOPHOTONICS 2018; 11:e201700329. [PMID: 29704320 DOI: 10.1002/jbio.201700329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/26/2018] [Indexed: 06/08/2023]
Abstract
Nanosecond pulsed laser irradiation can trigger a release of nucleic acids from gold nanoparticles, but the involved nanoeffects are not fully understood yet. Here we investigate the release of coumarin labeled siRNA from 15 to 30 nm gold particles after nanosecond pulsed laser irradiation. Temperatures in the particle and near the surface were calculated for the different radiant exposures. Upon irradiation with laser pulses of 4 nanosecond duration release started for both particle sizes at a calculated temperature increase of approximately 500 K. Maximum coumarin release was observed for 15 nm particles after irradiation with radiant exposure of 80 mJ cm-2 and with 32 mJ cm-2 for 30 nm particles. This corresponds to a temperature increase of 815 and 900 K, respectively. Our results show that the molecular release by nanosecond pulsed irradiation is based on a different mechanism compared to continuous or femtosecond irradiation. Local temperatures are considerably higher and it is expected that bubble formation plays a crucial role in release and damage to cellular structures.
Collapse
Affiliation(s)
- Florian Rudnitzki
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany
| | - Susanne Feineis
- Department and Chair of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | | | - Elmar Endl
- Institutes of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Johanna Lutz
- Department and Chair of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | - Jürgen Groll
- Department and Chair of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | - Gereon Hüttmann
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany
- Medizinisches Laserzentrum Lübeck GmbH, Lübeck, Germany
| |
Collapse
|
219
|
Liu T, Shi C, Duan L, Zhang Z, Luo L, Goel S, Cai W, Chen T. A highly hemocompatible erythrocyte membrane-coated ultrasmall selenium nanosystem for simultaneous cancer radiosensitization and precise antiangiogenesis. J Mater Chem B 2018; 6:4756-4764. [PMID: 30450208 PMCID: PMC6234506 DOI: 10.1039/c8tb01398e] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Radiotherapy is a vitally important strategy for clinical treatment of malignant cancers. Therefore, rational design and development of radiosensitizers that could enhance radiotherapeutic efficacy has attracted tremendous attention. Antiangiogenesis therapy could be a potentially effective strategy to regulate tumor growth and metastasis due to angiogenesis plays a pivotal role for tumor growth, invasion and metastasis to other organs. Herein, we have rationally designed a smart and effective nanosystem by combining ultrasmall selenium nanoparticles and bevacizumab (Avastin™, Av), for simultaneous radiotherapy and antiangiogenic therapy of cancer. The nanosystem was further coated with red blood cell (RBC) membranes to develop the final construct, RBCs@Se/Av. The RBC membrane coating effectively prolongs the blood circulation time and reduces the elimination of the nanosystem by autoimmune responses. As expected, RBCs@Se/Av, when irradiated with X-ray demonstrated potent anticancer and antiangiogenesis response in vitro and in vivo, as evidenced by strong inhibition of A375 tumor growth in nude mice, without causing any obvious histological damage to the non-target major organs. Taken together, this study demonstrates an effective strategy for design of smart Se-based nanosystem decorated with RBC membrane for simultaneous cancer radiosensitization and precise antiangiogenesis.
Collapse
Affiliation(s)
- Ting Liu
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Changzheng Shi
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Linqi Duan
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Zehang Zhang
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Liangping Luo
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Shreya Goel
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI 53705, USA
| | - Tianfeng Chen
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
220
|
Abstract
Nanoparticle delivery systems offer advantages over free drugs, in that they increase solubility and biocompatibility. Nanoparticles can deliver a high payload of therapeutic molecules while limiting off-target side effects. Therefore, delivery of an existing drug with a nanoparticle frequently results in an increased therapeutic index. Whether of synthetic or biologic origin, nanoparticle surface coatings are often required to reduce immune clearance and thereby increase circulation times allowing the carriers to reach their target site. To this end, polyethylene glycol (PEG) has long been used, with several PEGylated products reaching clinical use. Unfortunately, the growing use of PEG in consumer products has led to an increasing prevalence of PEG-specific antibodies in the human population, which in turn has fueled the search for alternative coating strategies. This review highlights alternative bioinspired nanoparticle shielding strategies, which may be more beneficial moving forward than PEG and other synthetic polymer coatings.
Collapse
Affiliation(s)
- Neetu M. Gulati
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Phoebe L. Stewart
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Radiology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Materials Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Macromolecular Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
221
|
Li B, Wang F, Gui L, He Q, Yao Y, Chen H. The potential of biomimetic nanoparticles for tumor-targeted drug delivery. Nanomedicine (Lond) 2018; 13:2099-2118. [DOI: 10.2217/nnm-2018-0017] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Bowen Li
- Department of Bioengineering, University of Washington, Seattle, Washington WA 98195, USA
| | - Fei Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Lijuan Gui
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Qing He
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Yuxin Yao
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, PR China
| |
Collapse
|
222
|
Liang H, Huang K, Su T, Li Z, Hu S, Dinh PU, Wrona EA, Shao C, Qiao L, Vandergriff AC, Hensley MT, Cores J, Allen T, Zhang H, Zeng Q, Xing J, Freytes DO, Shen D, Yu Z, Cheng K. Mesenchymal Stem Cell/Red Blood Cell-Inspired Nanoparticle Therapy in Mice with Carbon Tetrachloride-Induced Acute Liver Failure. ACS NANO 2018; 12:6536-6544. [PMID: 29943967 PMCID: PMC6373867 DOI: 10.1021/acsnano.8b00553] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Acute liver failure is a critical condition characterized by global hepatocyte death and often time needs a liver transplantation. Such treatment is largely limited by donor organ shortage. Stem cell therapy offers a promising option to patients with acute liver failure. Yet, therapeutic efficacy and feasibility are hindered by delivery route and storage instability of live cell products. We fabricated a nanoparticle that carries the beneficial regenerative factors from mesenchymal stem cells and further coated it with the membranes of red blood cells to increase blood stability. Unlike uncoated nanoparticles, these particles promote liver cell proliferation in vitro and have lower internalization by macrophage cells. After intravenous delivery, these artificial stem cell analogs are able to remain in the liver and mitigate carbon tetrachloride-induced liver failure in a mouse model, as gauged by histology and liver function test. Our technology provides an innovative and off-the-shelf strategy to treat liver failure.
Collapse
Affiliation(s)
- Hongxia Liang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Analytical Chemistry Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, Hebei 071002, China
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Phuong-Uyen Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Emily A. Wrona
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Chen Shao
- Department of Pathology, China-Japan Friendship Hospital, Peking 100029, China
| | - Li Qiao
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Adam C. Vandergriff
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - M. Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Tyler Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Hongyu Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qinglei Zeng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jiyuan Xing
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Deliang Shen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zujiang Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Corresponding Authors:,
| | - Ke Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
- Corresponding Authors:,
| |
Collapse
|
223
|
Gao C, Lin Z, Lin X, He Q. Cell Membrane-Camouflaged Colloid Motors for Biomedical Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800056] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Changyong Gao
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Zhihua Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Xiankun Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Qiang He
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| |
Collapse
|
224
|
Chen Y, Chen M, Zhang Y, Lee JH, Escajadillo T, Gong H, Fang RH, Gao W, Nizet V, Zhang L. Broad-Spectrum Neutralization of Pore-Forming Toxins with Human Erythrocyte Membrane-Coated Nanosponges. Adv Healthc Mater 2018; 7:e1701366. [PMID: 29436150 PMCID: PMC6041168 DOI: 10.1002/adhm.201701366] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/15/2018] [Indexed: 01/01/2023]
Abstract
Neutralization of bacterial toxins has become a compelling approach to treating bacterial infections as it may pose less selective pressure for the development of bacterial resistance. Currently, the majority of toxin neutralization platforms act by targeting the molecular structure of the toxin, which requires toxin identification and customized design for different diseases. Therefore, their development has been challenged by the enormous number and complexity of bacterial toxins. Herein, biomimetic toxin nanosponges are formulated by coating membranes of human red blood cells (hRBCs) onto polymeric nanoparticles, which act as a toxin decoy to absorb and neutralize a broad-spectrum of hemolytic toxins regardless of their molecular structure. When tested with model pore-forming toxins, including melittin, α-hemolysin of methicillin-resistant Staphylococcus aureus, listeriolysin O of Listeria monocytogenes, and streptolysin O of Group A Streptococcus, the hRBC nanosponges are able to completely inhibit toxin-induced hemolysis in a concentration-dependent manner. In addition, the nanosponge-detained toxins show no cytotoxicity when tested on human umbilical vein endothelial cells and no lethality when injected into mice, which together indicate effective toxin neutralization. Overall, these results demonstrate the broad applicability and high effectiveness of the hRBC nanosponges as a novel antivirulence platform against hemolytic toxins from various strains of bacteria.
Collapse
Affiliation(s)
- Yijie Chen
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Mengchun Chen
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yue Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Joo Hee Lee
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Tamara Escajadillo
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, USA
| | - Hua Gong
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
225
|
Gao S, Zheng P, Li Z, Feng X, Yan W, Chen S, Guo W, Liu D, Yang X, Wang S, Liang XJ, Zhang J. Biomimetic O 2-Evolving metal-organic framework nanoplatform for highly efficient photodynamic therapy against hypoxic tumor. Biomaterials 2018; 178:83-94. [PMID: 29913389 DOI: 10.1016/j.biomaterials.2018.06.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/18/2022]
Abstract
Improving the supply of O2 and the circulation lifetime of photosensitizers for photodynamic therapy (PDT) in vivo would be a promising approach to eliminate hypoxic tumors. Herein, by taking advantage of the significant gas-adsorption capability of metal-organic frameworks (MOFs), a biomimetic O2-evolving photodynamic therapy (PDT) nanoplatform with long circulating properties was fabricated. Zirconium (IV)-based MOF (UiO-66) was used as a vehicle for O2 storing, then conjugated with indocyanine green (ICG) by coordination reaction, and further coated with red blood cell (RBC) membranes. Upon 808 nm laser irradiation, the initial singlet oxygen (1O2) generated by ICG would decompose RBC membranes. At the same time, The photothermal property of ICG could facilitate the burst release of O2 from UiO-66. Subsequently, the generated O2 could significantly improve the PDT effects on hypoxic tumor. Owing to the advantages of long circulation and O2 self-sufficient, the designed nanotherapeutic agent can improve the efficiency of treatment against hypoxia tumor via PDT. Hence, this study presents a new paradigm for co-delivery of O2 and photosensitizers, and provides a new avenue to eliminate hypoxic tumors.
Collapse
Affiliation(s)
- Shutao Gao
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China; College of Science, Agricultural University of Hebei, Baoding, 071001, PR China
| | - Pengli Zheng
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Zhenhua Li
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China; Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.
| | - Xiaochen Feng
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Weixiao Yan
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Shizhu Chen
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China; CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, PR China
| | - Weisheng Guo
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, PR China
| | - Dandan Liu
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Xinjian Yang
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Shuxiang Wang
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, PR China.
| | - Jinchao Zhang
- College of Chemistry& Environmental Science, Analytical Chemistry Key Laboratory of Hebei Province, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, PR China.
| |
Collapse
|
226
|
Liu X, Li X, Xu W, Zhang X, Huang Z, Wang F, Liu J. Sub-Angstrom Gold Nanoparticle/Liposome Interfaces Controlled by Halides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:6628-6635. [PMID: 29741377 DOI: 10.1021/acs.langmuir.8b01138] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A hallmark of nanoscience is size-dependent and distance-dependent physical properties. Although most previous studies focused on optical properties, which are often tuned at nanometer scale, we herein report on the interaction between halide-capped gold nanoparticles (AuNPs) and phosphocholine (PC) liposomes at the sub-Angstrom level. Halide-capped AuNPs are adsorbed by PC liposomes attributable to van der Waals force. Iodide-capped AuNPs interact much more weakly with the liposomes compared with bromide- and chloride-capped AuNPs, as indicated by a liposome leakage assay and differential scanning calorimetry. This is explained by the slightly larger size of iodide separating the AuNP core more from the liposome surface. Cryo-transmission electron microscopy indicates that the liposomes remain intact when mixed with these halide-capped AuNPs of 13 or 70 nm in diameter. Other even larger ligands, including small thiol compounds, DNA oligonucleotides, proteins, and polymers, fully blocked the interaction, whereas AuNPs dispersed in noninteracting ions, including fluoride, phosphate, perchlorate, nitrate, sulfate, and bicarbonate, are still adsorbed strongly by 1,2-dioleoyl- sn-glycero-3-phosphocholine liposomes. Taken together, halides can be used to control interparticle distances at an extremely small scale with remarkable effects on materials properties, allowing surface probing, biosensor development, and fundamental surface science studies.
Collapse
Affiliation(s)
- Xiuru Liu
- School of Biological and Medical Engineering , Hefei University of Technology , Hefei , Anhui 230009 , China
| | - Xiaoqiu Li
- Center of Intervention Radiology, Center of Precise Medicine , Zhuhai People's Hospital , No. 79 Kangning Road , Zhuhai , Guangdong Province 519000 , China
| | - Wu Xu
- School of Biological and Medical Engineering , Hefei University of Technology , Hefei , Anhui 230009 , China
| | - Xiaohan Zhang
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| | - Zhicheng Huang
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| | - Feng Wang
- School of Biological and Medical Engineering , Hefei University of Technology , Hefei , Anhui 230009 , China
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| |
Collapse
|
227
|
Charoenphol P, Oswalt K, Bishop CJ. Therapeutics incorporating blood constituents. Acta Biomater 2018; 73:64-80. [PMID: 29626699 DOI: 10.1016/j.actbio.2018.03.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/01/2018] [Accepted: 03/28/2018] [Indexed: 12/17/2022]
Abstract
Blood deficiency and dysfunctionality can result in adverse events, which can primarily be treated by transfusion of blood or the re-introduction of properly functioning sub-components. Blood constituents can be engineered on the sub-cellular (i.e., DNA recombinant technology) and cellular level (i.e., cellular hitchhiking for drug delivery) for supplementing and enhancing therapeutic efficacy, in addition to rectifying dysfunctioning mechanisms (i.e., clotting). Herein, we report the progress of blood-based therapeutics, with an emphasis on recent applications of blood transfusion, blood cell-based therapies and biomimetic carriers. Clinically translated technologies and commercial products of blood-based therapeutics are subsequently highlighted and perspectives on challenges and future prospects are discussed. STATEMENT OF SIGNIFICANCE Blood-based therapeutics is a burgeoning field and has advanced considerably in recent years. Blood and its constituents, with and without modification (i.e., combinatorial), have been utilized in a broad spectrum of pre-clinical and clinically-translated treatments. This review article summarizes the most up-to-date progress of blood-based therapeutics in the following contexts: synthetic blood substitutes, acellular/non-recombinant therapies, cell-based therapies, and therapeutic sub-components. The article subsequently discusses clinically-translated technologies and future prospects thereof.
Collapse
|
228
|
Fang RH, Kroll AV, Gao W, Zhang L. Cell Membrane Coating Nanotechnology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30. [PMID: 29582476 PMCID: PMC5984176 DOI: 10.1002/adma.201706759,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nanoparticle-based therapeutic, prevention, and detection modalities have the potential to greatly impact how diseases are diagnosed and managed in the clinic. With the wide range of nanomaterials available, the rational design of nanocarriers on an application-specific basis has become increasingly commonplace. Here, a comprehensive overview is provided on an emerging platform: cell-membrane-coating nanotechnology. As a fundamental unit of biology, cells carry out a wide range of functions, including the remarkable ability to interface and interact with their surrounding environment. Instead of attempting to replicate such functions via synthetic techniques, researchers are now directly leveraging naturally derived cell membranes as a means of bestowing nanoparticles with enhanced biointerfacing capabilities. This top-down technique is facile, highly generalizable, and has the potential to greatly augment existing nanocarriers. Further, the introduction of a natural membrane substrate onto nanoparticles surfaces has enabled additional applications beyond those traditionally associated with nanomedicine. Despite its relative youth, there exists an impressive body of literature on cell membrane coating, which is covered here in detail. Overall, there is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology.
Collapse
|
229
|
Fang RH, Kroll AV, Gao W, Zhang L. Cell Membrane Coating Nanotechnology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706759. [PMID: 29582476 PMCID: PMC5984176 DOI: 10.1002/adma.201706759] [Citation(s) in RCA: 1004] [Impact Index Per Article: 167.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/12/2017] [Indexed: 05/03/2023]
Abstract
Nanoparticle-based therapeutic, prevention, and detection modalities have the potential to greatly impact how diseases are diagnosed and managed in the clinic. With the wide range of nanomaterials available, the rational design of nanocarriers on an application-specific basis has become increasingly commonplace. Here, a comprehensive overview is provided on an emerging platform: cell-membrane-coating nanotechnology. As a fundamental unit of biology, cells carry out a wide range of functions, including the remarkable ability to interface and interact with their surrounding environment. Instead of attempting to replicate such functions via synthetic techniques, researchers are now directly leveraging naturally derived cell membranes as a means of bestowing nanoparticles with enhanced biointerfacing capabilities. This top-down technique is facile, highly generalizable, and has the potential to greatly augment existing nanocarriers. Further, the introduction of a natural membrane substrate onto nanoparticles surfaces has enabled additional applications beyond those traditionally associated with nanomedicine. Despite its relative youth, there exists an impressive body of literature on cell membrane coating, which is covered here in detail. Overall, there is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology.
Collapse
Affiliation(s)
- Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Ashley V. Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
230
|
Su J, Liu G, Lian Y, Kamal Z, Que X, Qiu Y, Qiu M. Preparation and characterization of erythrocyte membrane cloaked PLGA/arsenic trioxide nanoparticles and evaluation of their in vitro anti-tumor effect. RSC Adv 2018; 8:20068-20076. [PMID: 35541656 PMCID: PMC9080777 DOI: 10.1039/c8ra01417e] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/01/2018] [Indexed: 12/30/2022] Open
Abstract
Arsenic trioxide (ATO) is used for acute promyelocytic leukemia (APL) that is resistant to all-trans-retinoic acid, but its direct intravenous injection sometimes induces severe toxic side effects. Here, we developed a delivery system of red blood cell membrane (RBCM) cloaked poly (lactic-co-glycolic) acid (PLGA)-ATO nanoparticles (RPANs) to reduce the toxicity. PLGA was used to entrap the ATO, and the PLGA-ATO nanoparticles (PANs) were prepared by the emulsification method. Then RBCMs were employed to cloak the PANs using ultrasonication, to develop the RPANs delivery system. The prepared RPANs had a uniform size of around 233.6 nm with an obvious core-shell structure, as observed by TEM. The completeness of the membrane proteins was confirmed by SDS-PAGE and an in vitro release time of 65 h was determined for the RPANs. The RPANs also exhibited low cytotoxicity against the 293k kidney cell line (84.6% cell viability rate), which suggested that the ATO toxicity was reduced by RBCM cloaking. Moreover, the anti-tumor effects of the RPANs against the HL60 cell line were comparable to those of ATO solution. Our study demonstrated that the RPANs system has anti-tumor potential and could be developed into a safe and sustained release delivery system for ATO.
Collapse
Affiliation(s)
- Jing Su
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Geyi Liu
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Yumei Lian
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | - Zul Kamal
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
- Department of Pharmacy, Shaheed Benazir Bhutto University Sheringal Dir (Upper) 18000 Khyber Pakhtunkhwa Pakistan
| | - Xiao Que
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| | | | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University 800, Dongchuan Road Shanghai 200240 China +86-021-34204052 +86-021-34204052
| |
Collapse
|
231
|
Shao J, Abdelghani M, Shen G, Cao S, Williams DS, van Hest JCM. Erythrocyte Membrane Modified Janus Polymeric Motors for Thrombus Therapy. ACS NANO 2018; 12:4877-4885. [PMID: 29733578 PMCID: PMC5968433 DOI: 10.1021/acsnano.8b01772] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/07/2018] [Indexed: 05/20/2023]
Abstract
We report the construction of erythrocyte membrane-cloaked Janus polymeric motors (EM-JPMs) which are propelled by near-infrared (NIR) laser irradiation and are successfully applied in thrombus ablation. Chitosan (a natural polysaccharide with positive charge, CHI) and heparin (glycosaminoglycan with negative charge, Hep) were selected as wall materials to construct biodegradable and biocompatible capsules through the layer-by-layer self-assembly technique. By partially coating the capsule with a gold (Au) layer through sputter coating, a NIR-responsive Janus structure was obtained. Due to the asymmetric distribution of Au, a local thermal gradient was generated upon NIR irradiation, resulting in the movement of the JPMs through the self-thermophoresis effect. The reversible "on/off" motion of the JPMs and their motile behavior were easily tuned by the incident NIR laser intensity. After biointerfacing the Janus capsules with an erythrocyte membrane, the EM-JPMs displayed red blood cell related properties, which enabled them to move efficiently in relevant biological environments (cell culture, serum, and blood). Furthermore, this therapeutic platform exhibited excellent performance in ablation of thrombus through photothermal therapy. As man-made micromotors, these biohybrid EM-JPMs hold great promise of navigating in vivo for active delivery while overcoming the drawbacks of existing synthetic therapeutic platforms. We expect that this biohybrid motor has considerable potential to be widely used in the biomedical field.
Collapse
|
232
|
Burnouf T, Burnouf PA, Wu YW, Chuang EY, Lu LS, Goubran H. Circulatory-cell-mediated nanotherapeutic approaches in disease targeting. Drug Discov Today 2018; 23:934-943. [DOI: 10.1016/j.drudis.2017.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/15/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
|
233
|
Meng QF, Rao L, Zan M, Chen M, Yu GT, Wei X, Wu Z, Sun Y, Guo SS, Zhao XZ, Wang FB, Liu W. Macrophage membrane-coated iron oxide nanoparticles for enhanced photothermal tumor therapy. NANOTECHNOLOGY 2018; 29:134004. [PMID: 29334363 DOI: 10.1088/1361-6528/aaa7c7] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanotechnology possesses the potential to revolutionize the diagnosis and treatment of tumors. The ideal nanoparticles used for in vivo cancer therapy should have long blood circulation times and active cancer targeting. Additionally, they should be harmless and invisible to the immune system. Here, we developed a biomimetic nanoplatform with the above properties for cancer therapy. Macrophage membranes were reconstructed into vesicles and then coated onto magnetic iron oxide nanoparticles (Fe3O4 NPs). Inherited from the Fe3O4 core and the macrophage membrane shell, the resulting Fe3O4@MM NPs exhibited good biocompatibility, immune evasion, cancer targeting and light-to-heat conversion capabilities. Due to the favorable in vitro and in vivo properties, biomimetic Fe3O4@MM NPs were further used for highly effective photothermal therapy of breast cancer in nude mice. Surface modification of synthetic nanomaterials with biomimetic cell membranes exemplifies a novel strategy for designing an ideal nanoplatform for translational medicine.
Collapse
Affiliation(s)
- Qian-Fang Meng
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Kim I, Kwon D, Lee D, Lee TH, Lee JH, Lee G, Yoon DS. A highly permselective electrochemical glucose sensor using red blood cell membrane. Biosens Bioelectron 2018; 102:617-623. [DOI: 10.1016/j.bios.2017.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/22/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023]
|
235
|
Xuan M, Shao J, Zhao J, Li Q, Dai L, Li J. Magnetic Mesoporous Silica Nanoparticles Cloaked by Red Blood Cell Membranes: Applications in Cancer Therapy. Angew Chem Int Ed Engl 2018; 57:6049-6053. [DOI: 10.1002/anie.201712996] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/04/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
| | - Qi Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| | - Luru Dai
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| |
Collapse
|
236
|
Xuan M, Shao J, Zhao J, Li Q, Dai L, Li J. Magnetic Mesoporous Silica Nanoparticles Cloaked by Red Blood Cell Membranes: Applications in Cancer Therapy. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
| | - Qi Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| | - Luru Dai
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| |
Collapse
|
237
|
Ai X, Hu M, Wang Z, Zhang W, Li J, Yang H, Lin J, Xing B. Recent Advances of Membrane-Cloaked Nanoplatforms for Biomedical Applications. Bioconjug Chem 2018; 29:838-851. [PMID: 29509403 DOI: 10.1021/acs.bioconjchem.8b00103] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In terms of the extremely small size and large specific surface area, nanomaterials often exhibit unusual physical and chemical properties, which have recently attracted considerable attention in bionanotechnology and nanomedicine. Currently, the extensive usage of nanotechnology in medicine holds great potential for precise diagnosis and effective therapeutics of various human diseases in clinical practice. However, a detailed understanding regarding how nanomedicine interacts with the intricate environment in complex living systems remains a pressing and challenging goal. Inspired by the diversified membrane structures and functions of natural prototypes, research activities on biomimetic and bioinspired membranes, especially for those cloaking nanosized platforms, have increased exponentially. By taking advantage of the flexible synthesis and multiple functionality of nanomaterials, a variety of unique nanostructures including inorganic nanocrystals and organic polymers have been widely devised to substantially integrate with intrinsic biomoieties such as lipids, glycans, and even cell and bacteria membrane components, which endow these abiotic nanomaterials with specific biological functionalities for the purpose of detailed investigation of the complicated interactions and activities of nanomedicine in living bodies, including their immune response activation, phagocytosis escape, and subsequent clearance from vascular system. In this review, we summarize the strategies established recently for the development of biomimetic membrane-cloaked nanoplatforms derived from inherent host cells (e.g., erythrocytes, leukocytes, platelets, and exosomes) and invasive pathogens (e.g., bacteria and viruses), mainly attributed to their versatile membrane properties in biological fluids. Meanwhile, the promising biomedical applications based on nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective vaccine development for disease prevention, have also been outlined. Finally, the potential challenges and future prospects of the biomimetic membrane-cloaked nanoplatforms are also discussed.
Collapse
Affiliation(s)
- Xiangzhao Ai
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Ming Hu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Zhimin Wang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Wenmin Zhang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371.,College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Juan Li
- College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Huanghao Yang
- College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , China
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371.,College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| |
Collapse
|
238
|
Zhang Y, Cai K, Li C, Guo Q, Chen Q, He X, Liu L, Zhang Y, Lu Y, Chen X, Sun T, Huang Y, Cheng J, Jiang C. Macrophage-Membrane-Coated Nanoparticles for Tumor-Targeted Chemotherapy. NANO LETTERS 2018; 18:1908-1915. [PMID: 29473753 PMCID: PMC7470025 DOI: 10.1021/acs.nanolett.7b05263] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Various delivery vectors have been integrated within biologically derived membrane systems to extend their residential time and reduce their reticuloendothelial system (RES) clearance during systemic circulation. However, rational design is still needed to further improve the in situ penetration efficiency of chemo-drug-loaded membrane delivery-system formulations and their release profiles at the tumor site. Here, a macrophage-membrane-coated nanoparticle is developed for tumor-targeted chemotherapy delivery with a controlled release profile in response to tumor microenvironment stimuli. Upon fulfilling its mission of tumor homing and RES evasion, the macrophage-membrane coating can be shed via morphological changes driven by extracellular microenvironment stimuli. The nanoparticles discharged from the outer membrane coating show penetration efficiency enhanced by their size advantage and surface modifications. After internalization by the tumor cells, the loaded drug is quickly released from the nanoparticles in response to the endosome pH. The designed macrophage-membrane-coated nanoparticle (cskc-PPiP/PTX@Ma) exhibits an enhanced therapeutic effect inherited from both membrane-derived tumor homing and step-by-step controlled drug release. Thus, the combination of a biomimetic cell membrane and a cascade-responsive polymeric nanoparticle embodies an effective drug delivery system tailored to the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Chao Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xi He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lisha Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xinli Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Corresponding Author: ,
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Corresponding Author: ,
| |
Collapse
|
239
|
Li YJ, Yang CX, Yan XP. Biomimetic Persistent Luminescent Nanoplatform for Autofluorescence-Free Metastasis Tracking and Chemophotodynamic Therapy. Anal Chem 2018; 90:4188-4195. [PMID: 29504391 DOI: 10.1021/acs.analchem.8b00311] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Metastasis is the main cause of death in people with cancer. Early diagnosis and targeted therapy for metastasis is crucial for the survival of the cancer patients. However, metastasis is hard to trace for its small size, dispersed distribution and unvascularized anatomy. Here we report a biomimetic persistent luminescent nanoplatform for noninvasive high-sensitive diagnosis and 808 nm laser controlled photodynamics assisted chemotherapy of metastasis. The nanoplatform is composed of a photosensitizer functionalized persistent luminescent nanoparticle core, a doxorubicin loaded hollow silica interlayer and a cancer cell membrane shell for effective metastasis theranostic. The cancer cell membrane shell prevents drug leakage and endows the nanoplatform with targeting ability to metastasis. The reactivatable persistent luminescence of persistent luminescent nanoparticles not only enables long-term in vivo metastasis tracking, but also provides internal light source for singlet oxygen generation to kill cancer cells and further break the membrane shell for drug release. This work provides a promising strategy to develop persistent luminescence imaging guided theranostic nanoplatforms for early metastasis.
Collapse
Affiliation(s)
- Yu-Jie Li
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Cheng-Xiong Yang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Xiu-Ping Yan
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China.,State Key Laboratory of Food Science and Technology (Jiangnan University), Institute of Analytical Food Safety, School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China.,Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300071 , China
| |
Collapse
|
240
|
Yu B, Goel S, Ni D, Ellison PA, Siamof CM, Jiang D, Cheng L, Kang L, Yu F, Liu Z, Barnhart TE, He Q, Zhang H, Cai W. Reassembly of 89 Zr-Labeled Cancer Cell Membranes into Multicompartment Membrane-Derived Liposomes for PET-Trackable Tumor-Targeted Theranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704934. [PMID: 29430735 PMCID: PMC5878718 DOI: 10.1002/adma.201704934] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/03/2017] [Indexed: 05/02/2023]
Abstract
Nanoengineering of cell membranes holds great potential to revolutionize tumor-targeted theranostics, owing to their innate biocompatibility and ability to escape from the immune and reticuloendothelial systems. However, tailoring and integrating cell membranes with drug and imaging agents into one versatile nanoparticle are still challenging. Here, multicompartment membrane-derived liposomes (MCLs) are developed by reassembling cancer cell membranes with Tween-80, and are used to conjugate 89 Zr via deferoxamine chelator and load tetrakis(4-carboxyphenyl) porphyrin for in vivo noninvasive quantitative tracing by positron emission tomography imaging and photodynamic therapy (PDT), respectively. Radiolabeled constructs, 89 Zr-Df-MCLs, demonstrate excellent radiochemical stability in vivo, target 4T1 tumors by the enhanced permeability and retention effect, and are retained long-term for efficient and effective PDT while clearing gradually from the reticuloendothelial system via hepatobiliary excretion. Toxicity evaluation confirms that the MCLs do not impose acute or chronic toxicity in intravenously injected mice. Additionally, 89 Zr-labeled MCLs can execute rapid and highly sensitive lymph node mapping, even for deep-seated sentinel lymph nodes. The as-developed cell membrane reassembling route to MCLs could be extended to other cell types, providing a versatile platform for disease theranostics by facilely and efficiently integrating various multifunctional agents.
Collapse
Affiliation(s)
- Bo Yu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Shreya Goel
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dalong Ni
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul A Ellison
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Cerise M Siamof
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, China
| | - Lei Kang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Nuclear Medicine, Peking University First Hospital Beijing, Beijing, 100000, China
| | - Faquan Yu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, China
| | - Todd E Barnhart
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Qianjun He
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| |
Collapse
|
241
|
Pei Q, Hu X, Zheng X, Liu S, Li Y, Jing X, Xie Z. Light-Activatable Red Blood Cell Membrane-Camouflaged Dimeric Prodrug Nanoparticles for Synergistic Photodynamic/Chemotherapy. ACS NANO 2018; 12:1630-1641. [PMID: 29346736 DOI: 10.1021/acsnano.7b08219] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Biomimetic approach offers numerous opportunities to design therapeutic platforms with enhanced antitumor performance and biocompatibility. Herein we report red blood cell membrane-camouflaged nanoparticles (RBC(M(TPC-PTX))) for synergistic chemo- and photodynamic therapy (PDT). Specifically, the inner core is mainly constructed by reactive oxygen species (ROS)-responsive PTX dimer (PTX2-TK) and photosensitizer 5,10,15,20-tetraphenylchlorin (TPC). In vitro experiments show that the prepared RBC(M(TPC-PTX)) is readily taken up into endosomes. Under appropriate light irradiation, the TPC can generate ROS, not only for PDT but also for triggering PTX2-TK cleavage and on-demand PTX release for chemotherapy. In vivo results show that the coating of RBC membrane prolongs blood circulation and improves tumor accumulation. The combination of chemo- and photodynamic therapy enhances anticancer therapeutic activity, and light-triggered drug release reduces systematic toxicity. All these characteristics render the described technology extremely promising for cancer treatment.
Collapse
Affiliation(s)
- Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China , Hefei, Anhui 230026, P. R. China
| | - Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Xiaohua Zheng
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China , Hefei, Anhui 230026, P. R. China
| | - Shi Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Yawei Li
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
242
|
Lv Y, Liu M, Zhang Y, Wang X, Zhang F, Li F, Bao WE, Wang J, Zhang Y, Wei W, Ma G, Zhao L, Tian Z. Cancer Cell Membrane-Biomimetic Nanoprobes with Two-Photon Excitation and Near-Infrared Emission for Intravital Tumor Fluorescence Imaging. ACS NANO 2018; 12:1350-1358. [PMID: 29338190 DOI: 10.1021/acsnano.7b07716] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Biomimetic fluorescent nanoprobes capable of emitting near-infrared (NIR) fluorescence (λmax ≈ 720 nm) upon excitation of 800 nm light were developed. The key conjugated polymer enabled two-photon absorption and Förster resonance energy transfer (FRET) processes within the nanoprobes, which imparted the nanoprobes with ideal NIR-incoming-NIR-outgoing fluorescence features. The cancer cell membrane (CM) coating endowed these nanoprobes with perfect biocompatibility and highly specific targeting ability to homologous tumors. It was believed that CM encapsulation provided an additional protecting layer for the photoactive components residing in the core of nanoprobes for retaining their intrinsic fluorescing ability in the physiological milieu. The long-term structural integrity, excellent photostability (fluorescence decrease <10% upon 30 min illumination of 800 nm pulse laser), high NIR fluorescence quantum yield (∼20%), and long in vivo circulation time of the target nanoprobes were also confirmed. The ability of these feature-packed nanoprobes for circumventing the challenges of absorption and light scattering caused by cellular structures and tissues was definitely confirmed via in vivo and in vitro experiments. The superior performances of these nanoprobes in terms of fluorescence signaling as well as targeting specificity were verified in intravital fluorescence imaging on tumor-bearing model mice. Specifically, these nanoprobes unequivocally enabled high-resolution visualization of the fine heterogeneous architectures of intravital tumor tissue, which proclaims the great potential of this type of probe for high-contrast fluorescence detection of thick biological samples in practical applications.
Collapse
Affiliation(s)
- Yanlin Lv
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences , Beijing 100049, P. R. China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Ming Liu
- School of Materials Science and Engineering, Harbin Institute of Technology , Harbin 150001, P. R. China
- School of Materials Science and Engineering, Wuhan Institute of Technology , Wuhan 403052, P. R. China
| | - Yong Zhang
- School of Materials Science and Engineering, Harbin Institute of Technology , Harbin 150001, P. R. China
| | - Xuefei Wang
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Fan Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Wei-Er Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Jie Wang
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Yuanlin Zhang
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, CAS , Beijing 100190, P. R. China
| | - Liancheng Zhao
- School of Materials Science and Engineering, Harbin Institute of Technology , Harbin 150001, P. R. China
| | - Zhiyuan Tian
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| |
Collapse
|
243
|
Liu JM, Zhang DD, Fang GZ, Wang S. Erythrocyte membrane bioinspired near-infrared persistent luminescence nanocarriers for in vivo long-circulating bioimaging and drug delivery. Biomaterials 2018; 165:39-47. [PMID: 29501968 DOI: 10.1016/j.biomaterials.2018.02.042] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 01/03/2023]
Abstract
Combination of biological entities with functional nanostructure would produce the excellent systemic drug-delivery vehicles that possess the ability to cross the biological barriers. Herein, from a biomimetic point of view, erythrocyte membrane bioinspired optical nanocarrier is fabricated by integrating Red blood cell (RBC) membrane vesicle with near-infrared persistent luminescence nanophosphors (PLNPs). The triple-doped zinc gallogermanate nanostructures with super-long near-infrared persistent luminescence (ZGGO) are used as optical emission center, mesoporous silica coated on the PLNPs (ZGGO@mSiO2) is employed for drug delivery, and the RBC membrane vesicle is introduced for biomimetic functionalization to ensure the developed nanocarriers bypass macrophage uptake and systemic clearance. Owing to the coating of natural erythrocyte membrane along with membrane lipids and associated membrane proteins, the proposed bioinspired nanocarriers have exhibited cell-mimicking property. Retaining the applicability of PLNPs core that favored in vitro excitation, the developed RBC-ZGGO@mSiO2 biomimetic nanocarriers have demonstrated intense fluorescence, super-long persistent luminescence, monodispersed nanosize, red light renewability, and excellent biocompatibility. In vivo mice bioimaging and biodistribution study demonstrate the erythrocyte membrane bioinspired nanoprobe loaded with doxorubicin as ideal nanocarriers for long-circulating bioimaging, in situ real-time monitoring and drug delivery. We believe the PLNPs-based biomimetic nanocarriers offer a promising nano-platform for diagnostics and therapeutics application.
Collapse
Affiliation(s)
- Jing-Min Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Dong-Dong Zhang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Guo-Zhen Fang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
244
|
Tumor targeted, stealthy and degradable bismuth nanoparticles for enhanced X-ray radiation therapy of breast cancer. Biomaterials 2018; 154:24-33. [DOI: 10.1016/j.biomaterials.2017.10.048] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/19/2017] [Accepted: 10/29/2017] [Indexed: 01/18/2023]
|
245
|
Li Z, Peng H, Liu J, Tian Y, Yang W, Yao J, Shao Z, Chen X. Plant Protein-Directed Synthesis of Luminescent Gold Nanocluster Hybrids for Tumor Imaging. ACS APPLIED MATERIALS & INTERFACES 2018; 10:83-90. [PMID: 29220160 DOI: 10.1021/acsami.7b13088] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Nowadays, fluorescence detection has emerged as one of the most frequently used noninvasive biosensing methods to selectively monitor biological processes within living systems. Among fluorescent nanoparticles (NPs), gold nanoclusters (AuNCs) have been intensively studied because of their intrinsic fluorescence and their endowed biocompatible surface. Herein, we selected an abundant, low-cost, and sustainable plant protein, the pea protein isolate (PPI), for its excellent biocompatibility, biodegradability, and nonallergenic character to be employed as both a reducing and stabilizing agent to facilely produce AuNCs exhibiting a strong red fluorescence. Afterward, the formed AuNCs/PPI mixture was able to self-assemble into NPs (AuNCs/PPI NPs) with the size of about 100 nm simply through a dialyzing process. Taking advantage from the protein nature of PPI, AuNCs/PPI NPs demonstrate both excellent biocompatibility and colloidal stability. Moreover, AuNCs/PPI NPs showed a great capability when employed as a bioimaging probe for both in vitro and in vivo imaging. Finally, AuNCs/PPI NPs were coated with red blood cell (RBC) membranes to improve their blood circulation property and enhance their tumor enrichment ability to meet the requirement for practical use. Results convincingly show that such super NPs (RBC-coated AuNCs/PPI NPs) were able to successfully locate tumor in vivowith an excellent imaging capability, which provides a new strategy for bioimaging with fluorescent NPs.
Collapse
Affiliation(s)
- Zhao Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Haibao Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Jialin Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Ye Tian
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Jinrong Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Zhengzhong Shao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| | - Xin Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and ‡Laboratory of Advanced Materials, Fudan University , Shanghai 200433, P. R. China
| |
Collapse
|
246
|
Hou J, Cui L, Chen R, Xu X, Chen J, Yin L, Liu J, Shi Q, Yin J. Facile Fabrication of Hierarchically Thermoresponsive Binary Polymer Pattern for Controlled Cell Adhesion. Macromol Rapid Commun 2018; 39:e1700572. [DOI: 10.1002/marc.201700572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/27/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Jianwen Hou
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Lele Cui
- Polymer Materials Research Center; College of Materials Science and Chemical Engineering; Harbin Engineering University; Harbin 150001 P. R. China
| | - Runhai Chen
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Xiaodong Xu
- Polymer Materials Research Center; College of Materials Science and Chemical Engineering; Harbin Engineering University; Harbin 150001 P. R. China
| | - Jiayue Chen
- Wego Holding Company Limited; Weihai 264210 P. R. China
| | - Ligang Yin
- Wego Holding Company Limited; Weihai 264210 P. R. China
| | - Jingchuan Liu
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Jinghua Yin
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| |
Collapse
|
247
|
Li J, Angsantikul P, Liu W, Esteban-Fernández de Ávila B, Chang X, Sandraz E, Liang Y, Zhu S, Zhang Y, Chen C, Gao W, Zhang L, Wang J. Biomimetic Platelet-Camouflaged Nanorobots for Binding and Isolation of Biological Threats. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1704800. [PMID: 29193346 DOI: 10.1002/adma.201704800] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/29/2017] [Indexed: 05/18/2023]
Abstract
One emerging and exciting topic in robotics research is the design of micro-/nanoscale robots for biomedical operations. Unlike industrial robots that are developed primarily to automate routine and dangerous tasks, biomedical nanorobots are designed for complex, physiologically relevant environments, and tasks that involve unanticipated biological events. Here, a biologically interfaced nanorobot is reported, made of magnetic helical nanomotors cloaked with the plasma membrane of human platelets. The resulting biomimetic nanorobots possess a biological membrane coating consisting of diverse functional proteins associated with human platelets. Compared to uncoated nanomotors which experience severe biofouling effects and hence hindered propulsion in whole blood, the platelet-membrane-cloaked nanomotors disguise as human platelets and display efficient propulsion in blood over long time periods. The biointerfaced nanorobots display platelet-mimicking properties, including adhesion and binding to toxins and platelet-adhering pathogens, such as Shiga toxin and Staphylococcus aureus bacteria. The locomotion capacity and platelet-mimicking biological function of the biomimetic nanomotors offer efficient binding and isolation of these biological threats. The dynamic biointerfacing platform enabled by platelet-membrane cloaked nanorobots thus holds considerable promise for diverse biomedical and biodefense applications.
Collapse
Affiliation(s)
- Jinxing Li
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Pavimol Angsantikul
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Wenjuan Liu
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Xiaocong Chang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Elodie Sandraz
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yuyan Liang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Siyu Zhu
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yue Zhang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chuanrui Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joseph Wang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
248
|
Li J, Wang W, Zhang X, Yao H, Wei Z, Li X, Mu X, Jiang J, Zhang H. Seedless preparation of Au nanorods by hydroquinone assistant and red blood cell membrane camouflage. RSC Adv 2018; 8:21316-21325. [PMID: 35539950 PMCID: PMC9080879 DOI: 10.1039/c8ra03795g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/28/2018] [Indexed: 12/27/2022] Open
Abstract
Natural red blood cell membranes camouflaged Au nanorod composites that exhibited an excellent biocompatibility and photothermal ablation effect.
Collapse
Affiliation(s)
- Jing Li
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Wenjing Wang
- State Key Laboratory of Supramolecular Structure and Materials
- College of Chemistry
- Jilin University
- Changchun 130012
- P. R. China
| | - Xue Zhang
- College of Resources and Environment
- Jilin Agricultural University
- Changchun 130118
- China
| | - Hua Yao
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Zhenhong Wei
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Xiuying Li
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Xupeng Mu
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Jinlan Jiang
- The Scientific Research Center
- China-Japan Union Hospital
- Jilin University
- Changchun 130033
- P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials
- College of Chemistry
- Jilin University
- Changchun 130012
- P. R. China
| |
Collapse
|
249
|
Peng J, Yang Q, Li W, Tan L, Xiao Y, Chen L, Hao Y, Qian Z. Erythrocyte-Membrane-Coated Prussian Blue/Manganese Dioxide Nanoparticles as H 2O 2-Responsive Oxygen Generators To Enhance Cancer Chemotherapy/Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:44410-44422. [PMID: 29210279 DOI: 10.1021/acsami.7b17022] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Because of the nontargeting release of anticancer drugs, conventional chemotherapy results in serious side effects and poor therapeutic outcomes. In addition, hypoxia situation in the tumor microenvironment also promotes the growth and metastasis of tumors. Multifunctional nanocarriers with stimuli-activation and hypoxia-relieving properties can help overcome some of these limitations. In this study, we have constructed a nanocarrier which is named PBMn-DOX@RBC. A Prussian blue/manganese dioxide (PBMn) nanoparticle is used as an oxygen precursor or catalyzer for H2O2 activation, and a red blood cell (RBC) membrane is used to increase the loading capacity of doxorubicin (DOX) and prolong the circulation time in vivo. H2O2 is overproduced in tumor tissues and tumor cells. It can be used as a stimulus to activate drug release. In the presence of H2O2, the hypoxia inside the tumors is relieved by the administration of PBMn-DOX@RBC. The generated oxygen disrupts the RBC coated on the surface of PBMn, which accelerates the release of DOX. RBCs also prolong the circulation time of the nanometer system in vivo. By combining the photothermal therapy (PTT) and chemotherapy, the tumor growth inhibition mediated by PBMn-DOX@RBC is further enhanced. PBMn-DOX@RBC fulfills the demands to relieve tumor hypoxia and enhance cancer chemotherapy/PTT.
Collapse
Affiliation(s)
- Jinrong Peng
- State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , No. 17, Section 3, Southern Renmin Road, Chengdu, Sichuan, P. R. China
| | - Qian Yang
- School of Pharmacy, Chengdu Medical College , No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan, China
| | - Wenting Li
- Department of Pharmacy, West China Second University Hospital , Chengdu, Sichuan, P. R. China
| | - Liwei Tan
- Department of Pharmacy, West China Second University Hospital , Chengdu, Sichuan, P. R. China
| | - Yao Xiao
- State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , No. 17, Section 3, Southern Renmin Road, Chengdu, Sichuan, P. R. China
| | - Lijuan Chen
- State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , No. 17, Section 3, Southern Renmin Road, Chengdu, Sichuan, P. R. China
| | - Ying Hao
- State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , No. 17, Section 3, Southern Renmin Road, Chengdu, Sichuan, P. R. China
| | - Zhiyong Qian
- State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University , No. 17, Section 3, Southern Renmin Road, Chengdu, Sichuan, P. R. China
| |
Collapse
|
250
|
Zhang Z, Liu Y, Zhang X, Liu J. A Cell-Mimicking Structure Converting Analog Volume Changes to Digital Colorimetric Output with Molecular Selectivity. NANO LETTERS 2017; 17:7926-7931. [PMID: 29130302 DOI: 10.1021/acs.nanolett.7b04298] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We herein report a three-component cell-mimicking structure with a peroxidase-like iron oxide nanozyme as the nucleus, a molecularly imprinted hydrogel shell as cytoplasm, and a lipid bilayer membrane. The structure was characterized by cryo and negative stain TEM and also by a calcein leakage test. By introducing charged monomers, the gel shell can swell or shrink in response to salt concentration. By lowering the salt concentration, the gradual "analog" gel volume change was reflected in a switch-like "digital" colorimetric output by the burst of membrane and oxidation of substrates such as 3,3',5,5'-tetramethylbenzidine (TMB). Controlled access was also achieved by using melittin to insert channels cross the membrane, and selective molecular transport was realized by the molecularly imprinted gel. The functions of each component are coupled, and this sophisticated tripartite structure provides a new platform for modular design of new materials. Our cell-mimicking structure is functional and it is complementary to the current protocell work that aims to understand the origin of life.
Collapse
Affiliation(s)
- Zijie Zhang
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue W, Waterloo, Ontario N2L 3G1, Canada
| | - Yibo Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue W, Waterloo, Ontario N2L 3G1, Canada
| | - Xiaohan Zhang
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue W, Waterloo, Ontario N2L 3G1, Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue W, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|