101
|
Ma Q, Fan Q, Xu J, Bai J, Han X, Dong Z, Zhou X, Liu Z, Gu Z, Wang C. Calming Cytokine Storm in Pneumonia by Targeted Delivery of TPCA-1 Using Platelet-Derived Extracellular Vesicles. MATTER 2020; 3:287-301. [PMID: 32835220 PMCID: PMC7242942 DOI: 10.1016/j.matt.2020.05.017] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/03/2020] [Accepted: 05/16/2020] [Indexed: 05/18/2023]
Abstract
Pneumonia can cause high morbidity and mortality because of uncontrolled inflammation in the lung tissue. Calming the cytokine storm may be one key to saving the life of patients with severe pneumonia. Here, inspired by the intrinsic affinity of platelets to the site of inflammation, we have engineered platelet-derived extracellular vesicles (PEVs) for pneumonia-targeted drug delivery. It is demonstrated that PEVs that are easily generated from the activated platelets can selectively target pneumonia in the mouse model with acute lung injury (ALI). By loading with [5-(p-fluorophenyl)-2-ureido]thiophene-3-carboxamide (TPCA-1), which can inhibit the production of inflammatory factors, the PEVs significantly improve therapeutic benefits by inhibiting the infiltration of pulmonary inflammatory cells and calming local cytokine storm compared with the free drug-treated group. Furthermore, we find that PEVs could serve as a broad platform that can selectively target various inflammatory sites, including chronic atherosclerotic plaque, rheumatoid arthritis, and wounds associated with skin.
Collapse
Affiliation(s)
- Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qin Fan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jinyu Bai
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiao Han
- Department of Bioengineering and California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiaozhong Zhou
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhen Gu
- Department of Bioengineering and California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
102
|
Abdullah SM, Rashid H. Melatonin Ameliorates BPA Induced Oxidative Stress in Human Red Blood Cells: An In vitro Study. Endocr Metab Immune Disord Drug Targets 2020; 20:1321-1327. [PMID: 32368984 DOI: 10.2174/1871530320666200505112023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/14/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bisphenol A (BPA) is a xenobiotic that causes oxidative stress in various organs in living organisms. Blood cells are also an endpoint where BPA is known to cause oxidative stress. Blood cells, especially red blood cells (RBCs), are crucial for maintaining homeostasis and overall wellbeing of the organism. They are highly susceptible to oxidative stress induced by xenobiotics. However, there is limited data about the oxidative stress induced by BPA in blood, especially in red blood cells. This study was carried out to evaluate BPA induced oxidative stress in human RBCs in vitro and its amelioration by melatonin. OBJECTIVE To find if melatonin exerts a protective effect on the oxidative stress induced by the BPA in human red blood cells in vitro. METHODS The erythrocyte suspensions (2 ml) were divided into six groups and treated with 0, 50, 100, 150, 200, and 250 μg/ml of BPA. Another set of erythrocyte suspension with similar BPA treatment and 50 μM Melatonin per group was also set. Incubations lasted for 12 hrs in the dark. Lipid peroxidation, glutathione, glutathione reductase, catalase, and superoxide dismutase were measured as indicators of oxidative stress. RESULTS BPA caused a significant increase in lipid peroxidation. A decrease in GSH levels was also observed. The activities of all the studied antioxidants also decreased with BPA treatment. Melatonin was seen to mitigate the oxidative stress induced by BPA. CONCLUSION Treatment of red blood cells with BPA caused an increase in oxidative stress, while melatonin decreased the induced oxidative stress.
Collapse
Affiliation(s)
- Saleh M Abdullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hina Rashid
- Centre of Research for Development, University of Kashmir, Srinagar, 190 006, India
| |
Collapse
|
103
|
Zhang Y, Ma N, Luo C, Zhu J, Bao C. Photosensitizer-loaded cell membrane biomimetic nanoparticles for enhanced tumor synergetic targeted therapy. RSC Adv 2020; 10:9378-9386. [PMID: 35497215 PMCID: PMC9050061 DOI: 10.1039/c9ra08926h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/23/2020] [Indexed: 11/21/2022] Open
Abstract
Photodynamic therapy (PDT) has the advantages of low toxicity and specificity, but photosensitizers usually fail to accumulate efficiently at the tumor site. In this study, a new multifunctional nano-drug delivery system was exploited by a biomimetic strategy to improve the PDT effects. The self-assembled methoxy poly(ethylene glycol)-poly(lactide-co-glycolide) (mPEG-PLGA) nanoparticles encapsulated with the photosensitizer chlorin e6 (Ce6) by microfluidics were employed as the nano-core, followed by coating red blood cell (RBC) membranes as the biomimetic agent to prolong the circulation time in vivo. In order to boost the therapeutic effect, doxorubicin (Dox) was preloaded into RBC nanovesicles. The cell membrane surface was modified with folic acid (FA) to further enhance the tumor targeting efficiency. The prepared biomimetic nanoparticles with a homogeneous size (70 nm) can trigger sufficient reactive oxygen species (ROS), leading to significant tumor ablation without side effects. In addition, the system had high tumor targeting efficiency, with an increase of 25% compared with no FA-modified nanoparticles. Therefore, this biomimetic multifunctional nanodrug delivery system possesses a prolonged circulation time and higher tumor targeting efficiency and can exert better tumor cytotoxicity for improved PDT due to homophilic targeting in vivo.
Collapse
Affiliation(s)
- Yunjiao Zhang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Nan Ma
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Congcong Luo
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Jiaquan Zhu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Chunrong Bao
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| |
Collapse
|
104
|
Wang H, Williams GR, Xie X, Wu M, Wu J, Zhu LM. Stealth Polydopamine-Based Nanoparticles with Red Blood Cell Membrane for the Chemo-Photothermal Therapy of Cancer. ACS APPLIED BIO MATERIALS 2020; 3:2350-2359. [DOI: 10.1021/acsabm.0c00094] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Haijun Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Xiaotian Xie
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Meng Wu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Junzi Wu
- College of Basic Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
105
|
Tertis M, Cernat A, Mirel S, Cristea C. Nanodevices for Pharmaceutical and Biomedical Applications. ANAL LETT 2020. [DOI: 10.1080/00032719.2020.1728292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mihaela Tertis
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andreea Cernat
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Mirel
- Department of Medical Devices, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
106
|
Wang S, Yin Y, Song W, Zhang Q, Yang Z, Dong Z, Xu Y, Cai S, Wang K, Yang W, Wang X, Pang Z, Feng L. Red-blood-cell-membrane-enveloped magnetic nanoclusters as a biomimetic theranostic nanoplatform for bimodal imaging-guided cancer photothermal therapy. J Mater Chem B 2020; 8:803-812. [PMID: 31904076 DOI: 10.1039/c9tb01829h] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of red blood cell (RBC) membrane coatings has recently been found to be a biomimetic strategy to confer inner core nanomaterials with improved pharmacokinetic profiles by utilizing the intrinsic long blood circulation time of RBCs. Here, we envelope superparamagnetic nanoclusters (MNCs) with RBC membrane ghosts to obtain MNC@RBCs with significantly improved physiological stability compared to that of bare MNCs. After being loaded with near-infrared (NIR) cypate molecules, the as-prepared Cyp-MNC@RBCs show remarkably increased NIR absorbance and resultant efficient photothermal conversion efficacy. By tracking the NIR fluorescence of cypate in an in vivo fluorescence imaging system, we uncover that such Cyp-MNC@RBCs upon intravenous injection show significantly improved tumor-homing capacity as compared to bare cypate-loaded MNCs. A similar result is further evidenced by recording the T2-weighted magnetic resonance imaging (MRI) signal of MNCs. Furthermore, upon exposure to 808 nm laser irradiation, the tumors grown on the mice with the intravenous injection of Cyp-MNC@RBCs show a higher temperature increase than the tumors grown on the mice injected with plain MNC@RBCs and thus are significantly suppressed via photothermal ablation. This study presents the preparation of biomimetic Cyp-MNC@RBCs with greatly improved tumor-homing capacity as multifunctional nanotheranostic agents for fluorescence and MRI bimodal imaging-guided cancer photothermal therapy.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wang Song
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijuan Yang
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| | - Ziliang Dong
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Kuang Wang
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Wuli Yang
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Xuejun Wang
- Shanghai University of Traditional Chinese Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Liangzhu Feng
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| |
Collapse
|
107
|
Dong H, Xu X, Wang L, Mo R. Advances in living cell-based anticancer therapeutics. Biomater Sci 2020; 8:2344-2365. [DOI: 10.1039/d0bm00036a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review summarizes recent advances in the applications of living cells as drug carriers or active drugs for anticancer drug delivery and cancer therapy.
Collapse
Affiliation(s)
- He Dong
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Xiao Xu
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Leikun Wang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Ran Mo
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
108
|
Ji W, Smith PN, Koepsel RR, Andersen JD, Baker SL, Zhang L, Carmali S, Myerson JW, Muzykantov V, Russell AJ. Erythrocytes as carriers of immunoglobulin-based therapeutics. Acta Biomater 2020; 101:422-435. [PMID: 31669698 DOI: 10.1016/j.actbio.2019.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 11/25/2022]
Abstract
The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane in order to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. We developed four synthetic approaches to generate RBC-Staphylococcal protein A (RBC-SpA) complexes: amino group targeting through N-hydrosuccinidyl ester-functionalized homobifunctional poly(ethylene glycol) (NHS-PEG-NHS), direct thiol group targeting using heterobifunctional NHS-PEG-maleimide (NHS-PEG-MAL), converted thiol targeting using heterobifunctional NHS-PEG-MAL, and click chemistry using heterobifunctional NHS-PEG-azido (NHS-PEG-N3) and NHS-PEG-alkyne (NHS-PEG-alk). The RBC-PEG-SpA complexes were formed within minutes, followed by the attachment of over 105 antibodies per RBC to the accessible RBC-bound SpA via Fc-Protein A coupling. The RBC-PEG-SpA-antibody arrays were shown to be stable for more than 60 days in PBS and for more than 42 days in serum containing buffer. RBC-PEG-SpA-antibody complexes were shown to remove TNFα from physiological buffer and had similar mechanical properties to unmodified RBCs. Out of the four approaches, the converted thiol method provided the most controlled chemistry and construct stability. We are now ideally positioned to determine the long-term in vivo efficacy of chemically membrane-engineered RBCs to remove antigens, like TNFα, from serum. STATEMENT OF SIGNIFICANCE: The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. Conversion of RBCs into therapeutic delivery vehicles, we argue, would enhance the circulation life of immunoglobulin-based therapeutics while simultaneously evading deleterious immune response.
Collapse
Affiliation(s)
- Weihang Ji
- Disruptive Health Technology Institute, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Paige N Smith
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Richard R Koepsel
- Disruptive Health Technology Institute, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Jill D Andersen
- Disruptive Health Technology Institute, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Stefanie L Baker
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Libin Zhang
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Sheiliza Carmali
- Disruptive Health Technology Institute, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA
| | - Jacob W Myerson
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Muzykantov
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alan J Russell
- Disruptive Health Technology Institute, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA; Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA; Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA; Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA 15213, USA.
| |
Collapse
|
109
|
Liu L, He H, Liu J. Advances on Non-Genetic Cell Membrane Engineering for Biomedical Applications. Polymers (Basel) 2019; 11:E2017. [PMID: 31817418 PMCID: PMC6961000 DOI: 10.3390/polym11122017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapeutics are very promising modalities to address many unmet medical needs, including genetic engineering, drug delivery, and regenerative medicine as well as bioimaging. To enhance the function and improve the efficacy of cell-based therapeutics, a variety of cell surface engineering strategies (genetic engineering and non-genetic engineering) are developed to modify the surface of cells or cell-based therapeutics with some therapeutic molecules, artificial receptors, and multifunctional nanomaterials. In comparison to complicated procedures and potential toxicities associated with genetic engineering, non-genetic engineering strategies have emerged as a powerful and compatible complement to traditional genetic engineering strategies for enhancing the function of cells or cell-based therapeutics. In this review, we will first briefly summarize key non-genetic methodologies including covalent chemical conjugation (surface reactive groups-direct conjugation, and enzymatically mediated and metabolically mediated indirect conjugation) and noncovalent physical bioconjugation (biotinylation, electrostatic interaction, and lipid membrane fusion as well as hydrophobic insertion), which have been developed to engineer the surface of cell-based therapeutics with various materials. Next, we will comprehensively highlight the latest advances in non-genetic cell membrane engineering surrounding different cells or cell-based therapeutics, including whole-cell-based therapeutics, cell membrane-derived therapeutics, and extracellular vesicles. Advances will be focused specifically on cells that are the most popular types in this field, including erythrocytes, platelets, cancer cells, leukocytes, stem cells, and bacteria. Finally, we will end with the challenges, future trends, and our perspectives of this relatively new and fast-developing research field.
Collapse
Affiliation(s)
- Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
110
|
Liu Y, Luo J, Chen X, Liu W, Chen T. Cell Membrane Coating Technology: A Promising Strategy for Biomedical Applications. NANO-MICRO LETTERS 2019; 11:100. [PMID: 34138027 PMCID: PMC7770915 DOI: 10.1007/s40820-019-0330-9] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/14/2019] [Indexed: 05/02/2023]
Abstract
Cell membrane coating technology is an approach to the biomimetic replication of cell membrane properties, and is an active area of ongoing research readily applicable to nanoscale biomedicine. Nanoparticles (NPs) coated with cell membranes offer an opportunity to unite natural cell membrane properties with those of the artificial inner core material. The coated NPs not only increase their biocompatibility but also achieve effective and extended circulation in vivo, allowing for the execution of targeted functions. Although cell membrane-coated NPs offer clear advantages, much work remains before they can be applied in clinical practice. In this review, we first provide a comprehensive overview of the theory of cell membrane coating technology, followed by a summary of the existing preparation and characterization techniques. Next, we focus on the functions and applications of various cell membrane types. In addition, we collate model drugs used in cell membrane coating technology, and review the patent applications related to this technology from the past 10 years. Finally, we survey future challenges and trends pertaining to this technology in an effort to provide a comprehensive overview of the future development of cell membrane coating technology.
Collapse
Affiliation(s)
- Yao Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Jingshan Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Wei Liu
- 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.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
111
|
Dai X, Du T, Han K. Engineering Nanoparticles for Optimized Photodynamic Therapy. ACS Biomater Sci Eng 2019; 5:6342-6354. [DOI: 10.1021/acsbiomaterials.9b01251] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Xinxin Dai
- College of Science, Huazhong Agricultural University, No. 1 Shizishan Street, Wuhan 430070, China
| | - Ting Du
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin 300457, China
| | - Kai Han
- College of Science, Huazhong Agricultural University, No. 1 Shizishan Street, Wuhan 430070, China
- College of Pharmacy, University of Michigan, 2800 Plymouth Road, Ann Arbor, Michigan 48105, United States
| |
Collapse
|
112
|
Xu X, Li T, Shen S, Wang J, Abdou P, Gu Z, Mo R. Advances in Engineering Cells for Cancer Immunotherapy. Am J Cancer Res 2019; 9:7889-7905. [PMID: 31695806 PMCID: PMC6831467 DOI: 10.7150/thno.38583] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy aims to utilize the host immune system to kill cancer cells. Recent representative immunotherapies include T-cell transfer therapies, such as chimeric antigen receptor T cell therapy, antibody-based immunomodulator therapies, such as immune checkpoint blockade therapy, and cytokine therapies. Recently developed therapies leveraging engineered cells for immunotherapy against cancers have been reported to enhance antitumor efficacy while reducing side effects. Such therapies range from biologically, chemically and physically -engineered cells to bioinspired and biomimetic nanomedicines. In this review, advances of engineering cells for cancer immunotherapy are summarized, and prospects of this field are discussed.
Collapse
|
113
|
Han X, Shen S, Fan Q, Chen G, Archibong E, Dotti G, Liu Z, Gu Z, Wang C. Red blood cell-derived nanoerythrosome for antigen delivery with enhanced cancer immunotherapy. SCIENCE ADVANCES 2019; 5:eaaw6870. [PMID: 31681841 PMCID: PMC6810293 DOI: 10.1126/sciadv.aaw6870] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 09/16/2019] [Indexed: 05/04/2023]
Abstract
Erythrocytes or red blood cells (RBCs) represent a promising cell-mediated drug delivery platform due to their inherent biocompatibility. Here, we developed an antigen delivery system based on the nanoerythrosomes derived from RBCs, inspired by the splenic antigen-presenting cell targeting capacity of senescent RBCs. Tumor antigens were loaded onto the nanoerythrosomes by fusing tumor cell membrane-associated antigens with nanoerythrosomes. This tumor antigen-loaded nanoerythrosomes (nano-Ag@erythrosome) elicited antigen responses in vivo and, in combination with the anti-programmed death ligand 1 (PD-L1) blockade, inhibited the tumor growth in B16F10 and 4T1 tumor models. We also generated a tumor model showing that "personalized nano-Ag@erythrosomes" could be achieved by fusing RBCs and surgically removed tumors, which effectively reduced tumor recurrence and metastasis after surgery.
Collapse
Affiliation(s)
- Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shufang Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qin Fan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guojun Chen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Edikan Archibong
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gianpietro Dotti
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
114
|
Sonntag L, Simmchen J, Magdanz V. Nano-and Micromotors Designed for Cancer Therapy. Molecules 2019; 24:E3410. [PMID: 31546857 PMCID: PMC6767050 DOI: 10.3390/molecules24183410] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022] Open
Abstract
Research on nano- and micromotors has evolved into a frequently cited research area with innovative technology envisioned for one of current humanities' most deadly problems: cancer. The development of cancer targeting drug delivery strategies involving nano-and micromotors has been a vibrant field of study over the past few years. This review aims at categorizing recent significant results, classifying them according to the employed propulsion mechanisms starting from chemically driven micromotors, to field driven and biohybrid approaches. In concluding remarks of section 2, we give an insight into shape changing micromotors that are envisioned to have a significant contribution. Finally, we critically discuss which important aspects still have to be addressed and which challenges still lie ahead of us.
Collapse
Affiliation(s)
- Luisa Sonntag
- Chair of Physical Chemistry, TU Dresden, 01062 Dresden, Germany.
| | - Juliane Simmchen
- Chair of Physical Chemistry, TU Dresden, 01062 Dresden, Germany.
| | | |
Collapse
|
115
|
Zumbuehl A. Artificial Phospholipids and Their Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10223-10232. [PMID: 30278137 DOI: 10.1021/acs.langmuir.8b02601] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Phospholipids are at the heart and origin of life on this planet. The possibilities in terms of phospholipid self-assembly and biological functions seem limitless. Nonetheless, nature exploits only a small fraction of the available chemical space of phospholipids. Using chemical synthesis, artificial phospholipid structures become accessible, and the study of their biophysics may reveal unprecedented properties. In this article, the recent advances by our work group in the field of chemical lipidology are summarized. The family of diamidophospholipids is discussed in detail from monolayer characterization to the formation of faceted vesicles, culminating in the template-free self-assembly of phospholipid cubes and the possible applications of vesicle origami in modern personalized medicine.
Collapse
Affiliation(s)
- Andreas Zumbuehl
- Department of Chemistry , University of Fribourg , Chemin du Musée 9 , 1700 Fribourg , Switzerland
| |
Collapse
|
116
|
Debayle M, Marchandier T, Xu X, Lequeux N, Pons T. pH-Sensitive Visible or Shortwave Infrared Quantum Dot Nanoprobes Using Conformation-Switchable Copolymeric Ligands. ACS APPLIED MATERIALS & INTERFACES 2019; 11:25008-25016. [PMID: 31264837 DOI: 10.1021/acsami.9b06194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Intracellular and extracellular pH are key parameters in many physiological processes and diseases. For example, the extracellular pH of the tumor micro-environment is slightly more acidic than in healthy tissue. In vivo mapping of the extracellular pH within the tumor would therefore improve our understanding of the tumor physiology. Fluorescent semiconductor quantum dots (QDs) represent interesting probes for in vivo imaging, in particular in the shortwave infrared (SWIR) range. Here, pH-sensitive QD nanoprobes are developed using a conformation-switchable surface chemistry. The central fluorescent QD is coated with a copolymer ligand and conjugated to gold nanoparticle quenchers. As the pH decreases from physiological (7.5) to slightly acidic (5.5-6), the copolymer reversibly shrinks, which increases the energy transfer between the QD and the gold quenchers and modulates the QD fluorescence signal. This enables the design of ratiometric QD probes for biological pH range emitting in the visible or SWIR range. In addition, these probes can be easily encapsulated and remain functional within ghost erythrocyte membranes, which facilitate their in vivo application.
Collapse
Affiliation(s)
- Manon Debayle
- LPEM, UMR 8213, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université , 75005 Paris , France
| | - Thomas Marchandier
- LPEM, UMR 8213, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université , 75005 Paris , France
| | - Xiangzhen Xu
- LPEM, UMR 8213, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université , 75005 Paris , France
| | - Nicolas Lequeux
- LPEM, UMR 8213, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université , 75005 Paris , France
| | - Thomas Pons
- LPEM, UMR 8213, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université , 75005 Paris , France
| |
Collapse
|
117
|
Platelets as platforms for inhibition of tumor recurrence post-physical therapy by delivery of anti-PD-L1 checkpoint antibody. J Control Release 2019; 304:233-241. [DOI: 10.1016/j.jconrel.2019.05.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 04/29/2019] [Accepted: 05/04/2019] [Indexed: 12/24/2022]
|
118
|
Halder A, Sun Y. Biocompatible propulsion for biomedical micro/nano robotics. Biosens Bioelectron 2019; 139:111334. [PMID: 31128479 DOI: 10.1016/j.bios.2019.111334] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022]
Abstract
Micro/Nano robots have shown enormous potential for diverse biomedical applications, such as targeted delivery, in vivo biosensing, minimally invasive surgery and cell manipulation through extending their area of operation to various previously inaccessible locations. The motion of these small-scale robots can be either self-propelled or remotely controlled by some external power sources. However, in order to use them for biomedical applications, optimization of biocompatible propulsion and precise controllability are highly desirable. In this article, the recent progress about the biocompatible propulsion (e.g. self-propulsion, external stimuli based propulsion and bio-hybrid propulsion) techniques for these micro/nano robotic devices are summarized along with their applications, with a special focus on the advantages and disadvantages of different propulsion techniques. The current challenges and future perspectives of these small-scale devices are discussed in the final section.
Collapse
Affiliation(s)
- Arnab Halder
- Department of Health Technology, Technical University of Denmark, DK-2800, Kgs Lyngby, Denmark.
| | - Yi Sun
- Department of Health Technology, Technical University of Denmark, DK-2800, Kgs Lyngby, Denmark.
| |
Collapse
|
119
|
Wang H, Wu J, Williams GR, Fan Q, Niu S, Wu J, Xie X, Zhu LM. Platelet-membrane-biomimetic nanoparticles for targeted antitumor drug delivery. J Nanobiotechnology 2019; 17:60. [PMID: 31084622 PMCID: PMC6513513 DOI: 10.1186/s12951-019-0494-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/04/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Nanoscale drug-delivery systems (DDSs) have great promise in tumor diagnosis and treatment. Platelet membrane (PLTM) biomimetic DDSs are expected to enhance retention in vivo and escape uptake by macrophages, as well as minimizing immunogenicity, attributing to the CD47 protein in PLTM sends "don't eat me" signals to macrophages. In addition, P-selectin is overexpressed on the PLTM, which would allow a PLTM-biomimetic DDS to specifically bind to the CD44 receptors upregulated on the surface of cancer cells. RESULTS In this study, porous nanoparticles loaded with the anti-cancer drug bufalin (Bu) were prepared from a chitosan oligosaccharide (CS)-poly(lactic-co-glycolic acid) (PLGA) copolymer. These were subsequently coated with platelet membrane (PLTM) to form PLTM-CS-pPLGA/Bu NPs. The PLTM-CS-pPLGA/Bu NPs bear a particle size of ~ 192 nm, and present the same surface proteins as the PLTM. Confocal microscopy and flow cytometry results revealed a greater uptake of PLTM-CS-pPLGA/Bu NPs than uncoated CS-pPLGA/Bu NPs, as a result of the targeted binding of P-selectin on the surface of the PLTM to the CD44 receptors of H22 hepatoma cells. In vivo biodistribution studies in H22-tumor carrying mice revealed that the PLTM-CS-pPLGA NPs accumulated in the tumor, because of a combination of active targeting effect and the EPR effect. The PLTM-CS-pPLGA/Bu NPs led to more effective tumor growth inhibition over other bufalin formulations. CONCLUSIONS Platelet membrane biomimetic nanoparticles played a promising targeted treatment of cancer with low side effect.
Collapse
Affiliation(s)
- Haijun Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620 China
| | - Junzi Wu
- College of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, 650500 China
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX UK
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, 250117 China
| | - Shiwei Niu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620 China
| | - Jianrong Wu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620 China
| | - Xiaotian Xie
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620 China
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620 China
| |
Collapse
|
120
|
Xuan M, Shao J, Li J. Cell membrane-covered nanoparticles as biomaterials. Natl Sci Rev 2019; 6:551-561. [PMID: 34691904 PMCID: PMC8291551 DOI: 10.1093/nsr/nwz037] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Surface engineering of synthetic carriers is an essential and important strategy for drug delivery in vivo. However, exogenous properties make synthetic nanosystems invaders that easily trigger the passive immune clearance mechanism, increasing the retention effect caused by the reticuloendothelial systems and bioadhesion, finally leading to low therapeutic efficacy and toxic effects. Recently, a cell membrane cloaking technique has been reported as a novel interfacing approach from the biological/immunological perspective, and has proved useful for improving the performance of synthetic nanocarriers in vivo. After cell membrane cloaking, nanoparticles not only acquire the physiochemical properties of natural cell membranes but also inherit unique biological functions due to the presence of membrane-anchored proteins, antigens, and immunological moieties. The derived biological properties and functions, such as immunosuppressive capability, long circulation time, and targeted recognition integrated in synthetic nanosystems, have enhanced their potential in biomedicine in the future. Here, we review the cell membrane-covered nanosystems, highlight their novelty, introduce relevant biomedical applications, and describe the future prospects for the use of this novel biomimetic system constructed from a combination of cell membranes and synthetic nanomaterials.
Collapse
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| |
Collapse
|
121
|
Mu Q, Wang H, Gu X, Stephen ZR, Yen C, Chang FC, Dayringer CJ, Zhang M. Biconcave Carbon Nanodisks for Enhanced Drug Accumulation and Chemo-Photothermal Tumor Therapy. Adv Healthc Mater 2019; 8:e1801505. [PMID: 30856295 PMCID: PMC6483846 DOI: 10.1002/adhm.201801505] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/18/2019] [Indexed: 12/11/2022]
Abstract
It is considered a significant challenge to construct nanocarriers that have high drug loading capacity and can overcome physiological barriers to deliver efficacious amounts of drugs to solid tumors. Here, the development of a safe, biconcave carbon nanodisk to address this challenge for treating breast cancer is reported. The nanodisk demonstrates fluorescent imaging capability, an exceedingly high loading capacity (947.8 mg g-1 , 94.78 wt%) for doxorubicin (DOX), and pH-responsive drug release. It exhibits a higher uptake rate by tumor cells and greater accumulation in tumors in a mouse model than its carbon nanosphere counterpart. In addition, the nanodisk absorbs and transforms near-infrared (NIR) light to heat, which enables simultaneous NIR-responsive drug release for chemotherapy and generation of thermal energy for tumor cell destruction. Notably, this NIR-activated dual therapy demonstrates a near complete suppression of tumor growth in a mouse model of triple-negative breast cancer when DOX-loaded nanodisks are administered systemically.
Collapse
Affiliation(s)
- Qingxin Mu
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Hui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
- The Anhui Key Laboratory of Condensed Matter Physics at Extreme Conditions, High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
| | - Xinyu Gu
- Department of Biochemistry, University of Washington Seattle, Washington, DC, 98195, USA
| | - Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Charles Yen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Christopher J Dayringer
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| |
Collapse
|
122
|
Shen Y, Du K, Zou L, Zhou X, Lv R, Gao D, Qiu B, Ding W. Rapid and continuous on-chip loading of trehalose into erythrocytes. Biomed Microdevices 2019; 21:5. [PMID: 30607639 DOI: 10.1007/s10544-018-0352-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Freeze-drying is a promising approach for the long-term storage of erythrocytes at room temperature. Studies have shown that trehalose loaded into erythrocytes plays an important role in protecting erythrocytes against freeze-drying damage. Due to the impermeability of the erythrocyte membrane to trehalose, many methods have been developed to load trehalose into erythrocytes. However, these methods usually require multistep manual manipulation and long processing time; the adopted protocols are also diverse and not standardized. Thus, we develop an osmotically-based trehalose-loading microdevice (TLM) to rapidly, continuously, and automatically produce erythrocytes with loaded trehalose. In the TLM, trehalose is loaded through the erythrocyte membrane pores induced by hypotonic shock; then, the trehalose-loaded erythrocytes are rinsed to remove hemoglobin molecules and cell fragments, and the extracellular solution is restored to the isotonic state by integrating a rinsing-recovering design. First, the mixing function and the rinsing-recovering function were confirmed using a fluorescent solution. Then, the performance of the TLM was evaluated under various operating conditions with respect to the loading efficiency of trehalose, the hemolysis rate of erythrocytes (ϕ), the recovery rate of hemoglobin in erythrocytes (φ), and the separation efficiency of the TLM. Finally, the preliminary study of the freeze-drying of erythrocytes with loaded trehalose was accomplished using the TLM. The results showed that under the designated operating conditions, the loading efficiency for human erythrocytes reached ~21 mM in ~2 min with a ϕ value of ~17% and a φ value of ~74%. This study provides insights into the design of the on-chip loading of trehalose into erythrocytes and promotes the automation of life science studies on biochips.
Collapse
Affiliation(s)
- Yiren Shen
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Kun Du
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Lili Zou
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Xiaoming Zhou
- School of Mechatronics Engineering, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Rong Lv
- Hefei Blood Center, Hefei, 230000, Anhui, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Bensheng Qiu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Weiping Ding
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
123
|
Zhang X, Qiu M, Guo P, Lian Y, Xu E, Su J. Autologous Red Blood Cell Delivery of Betamethasone Phosphate Sodium for Long Anti-Inflammation. Pharmaceutics 2018; 10:pharmaceutics10040286. [PMID: 30567356 PMCID: PMC6320894 DOI: 10.3390/pharmaceutics10040286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/30/2022] Open
Abstract
Although glucocorticoids are highly effective in treating various types of inflammation such as skin disease, rheumatic disease, and allergic disease, their application have been seriously limited for their high incidence of side effects, particularly in long term treatment. To improve efficacy and reduce side effects, we encapsulated betamethasone phosphate (BSP) into biocompatible red blood cells (RBCs) and explored its long acting-effect. BSP was loaded into rat autologous erythrocytes by hypotonic preswelling method, and the loading amount was about 2.5 mg/mL cells. In vitro, BSP loaded RBCs (BSP-RBCs) presented similar morphology, osmotic fragility to native RBCs (NRBCs). After the loading process, the loaded cells can maintain around 70% of Na+/K+-ATPase activity of natural cells. In vivo, a series of tests including survival, pharmacokinetics, and anti-inflammatory effect were carried out to examine the long-acting effect of BSP-RBCs. The results shown that the loaded cells could circulate in plasma for over nine days, the release of BSP can last for over seven days and the anti-inflammatory effect can still be observed on day 5 after injection. Totally, BSP-loaded autologous erythrocytes seem to be a promising sustained releasing delivery system with long anti-inflammatory effect.
Collapse
Affiliation(s)
- Xiumei Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Pengcheng Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yumei Lian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Enge Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
124
|
Li Z, Hu S, Cheng K. Platelets and their biomimetics for regenerative medicine and cancer therapies. J Mater Chem B 2018; 6:7354-7365. [PMID: 31372220 PMCID: PMC6675472 DOI: 10.1039/c8tb02301h] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Platelets, circulating blood cells derived from megakaryocytes, play a key role in various physical activities, including coagulation, hemostasis, the body's innate immune response, and cancer metastasis. By taking advantage of their key traits, researchers have developed strategies to exploit platelets and platelet-mimicking nanoassemblies to treat a number of conditions, including wounds, cancers, and bacterial infections. Compared to traditional polymer, lipsosome, and inorganic nanoparticles-based delivery systems, platelets and platelet-mimicking vehicles hold many advantages. Among these are their enhanced circulation time, their large volumes and surface areas for drug loading or conjugation, and their inherent ability to target some diseases. In this review, we will highlight the recent progress made in the development of disease-targeting platelets- and platelet-mimicking-vehicles as therapeutic platforms.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
125
|
Liu L, Zhang S, Zhao L, Gu Z, Duan G, Zhou B, Yang Z, Zhou R. Superior Compatibility of C 2 N with Human Red Blood Cell Membranes and the Underlying Mechanism. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1803509. [PMID: 30474237 DOI: 10.1002/smll.201803509] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/28/2018] [Indexed: 06/09/2023]
Abstract
The widespread use of nanomaterials, such as carbon based 2D nanomaterials, in biomedical applications, has been accompanied by a growing concern on their biocompatibility, and in particular, on how they may affect the integrity of cell membranes. Herein, the interactions between C2 N, a novel 2D nanomaterial, and human red blood cell membranes are explored using a combined experimental and theoretical approach. The experimental microscopies show that C2 N exerts a negligible hemolysis effect on the blood cells with a superior compatibility to their cell membranes, when compared with the control system, reduced graphene oxide (rGO), which is found to be highly hemolytic. The molecular dynamics simulations further reveal the underlying molecular mechanisms, which indicate that C2 N prefers to be adsorbed flat on the water-membrane interface. Interaction energy analyses demonstrate the crucial role of Coulombic contributions, originating from the unique electrostatic potential surface of C2 N, in preventing C2 N from penetrating into cell membranes. These findings indicate a high compatibility of C2 N with cell membranes, which may provide useful foundation for the future exploration of this 2D nanomaterial in related biomedical applications.
Collapse
Affiliation(s)
- Lu Liu
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Shitong Zhang
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Lin Zhao
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Zonglin Gu
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Guangxin Duan
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Bo Zhou
- School of Electronic Engineering, Chengdu Technological University, Chengdu, 611730, China
| | - Zaixing Yang
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
| | - Ruhong Zhou
- Institute of Quantitative Biology and Medicine, State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Jiangsu, 215123, China
- IBM Thomas J. Watson Research Center, Yorktown Heights, NY, 10598, USA
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
126
|
Calderón-Garcidueñas L, González-Maciel A, Reynoso-Robles R, Kulesza RJ, Mukherjee PS, Torres-Jardón R, Rönkkö T, Doty RL. Alzheimer's disease and alpha-synuclein pathology in the olfactory bulbs of infants, children, teens and adults ≤ 40 years in Metropolitan Mexico City. APOE4 carriers at higher risk of suicide accelerate their olfactory bulb pathology. ENVIRONMENTAL RESEARCH 2018; 166:348-362. [PMID: 29935448 DOI: 10.1016/j.envres.2018.06.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/12/2018] [Accepted: 06/12/2018] [Indexed: 05/28/2023]
Abstract
There is growing evidence that air pollution is a risk factor for a number of neurodegenerative diseases, most notably Alzheimer's (AD) and Parkinson's (PD). It is generally assumed that the pathology of these diseases arises only later in life and commonly begins within olfactory eloquent pathways prior to the onset of the classical clinical symptoms. The present study demonstrates that chronic exposure to high levels of air pollution results in AD- and PD-related pathology within the olfactory bulbs of children and relatively young adults ages 11 months to 40 years. The olfactory bulbs (OBs) of 179 residents of highly polluted Metropolitan Mexico City (MMC) were evaluated for AD- and alpha-synuclein-related pathology. Even in toddlers, hyperphosphorylated tau (hTau) and Lewy neurites (LN) were identified in the OBs. By the second decade, 84% of the bulbs exhibited hTau (48/57), 68% LNs and vascular amyloid (39/57) and 36% (21/57) diffuse amyloid plaques. OB active endothelial phagocytosis of red blood cell fragments containing combustion-derived nanoparticles (CDNPs) and the neurovascular unit damage were associated with myelinated and unmyelinated axonal damage. OB hTau neurites were associated mostly with pretangle stages 1a and 1b in subjects ≤ 20 years of age, strongly suggesting olfactory deficits could potentially be an early guide of AD pretangle subcortical and cortical hTau. APOE4 versus APOE3 carriers were 6-13 times more likely to exhibit OB vascular amyloid, neuronal amyloid accumulation, alpha-synuclein aggregates, hTau neurofibrillary tangles, and neurites. Remarkably, APOE4 carriers were 4.57 times more likely than non-carriers to die by suicide. The present findings, along with previous data that over a third of clinically healthy MMC teens and young adults exhibit low scores on an odor identification test, support the concept that olfactory testing may aid in identifying young people at high risk for neurodegenerative diseases. Moreover, results strongly support early neuroprotective interventions in fine particulate matter (PM2.5) and CDNP's exposed individuals ≤ 20 years of age, and the critical need for air pollution control.
Collapse
Affiliation(s)
| | | | | | - Randy J Kulesza
- Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | | | - Ricardo Torres-Jardón
- Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, 04310, Mexico
| | - Topi Rönkkö
- Aerosol Physics, Faculty of Natural Sciences, Tampere University of Technology, FI-33101 Tampere, Finland
| | - Richard L Doty
- Smell and Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 19104, USA
| |
Collapse
|
127
|
Gautam M, Poudel K, Yong CS, Kim JO. Prussian blue nanoparticles: Synthesis, surface modification, and application in cancer treatment. Int J Pharm 2018; 549:31-49. [PMID: 30053487 DOI: 10.1016/j.ijpharm.2018.07.055] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 12/17/2022]
Abstract
This review outlines recently developed Prussian blue nanoparticle (PB NPs)-based multimodal imaging-guided chemo-photothermal strategies for cancer diagnosis and treatment in order to provide insight into the future of the field. The primary limitation of existing therapeutics is the lack of selectivity in drug delivery: they target healthy and cancerous cells alike. In this paper, we provide a thorough review of diverse synthetic and surface engineering techniques for PB NP fabrication. We have elucidated the various targeting approaches employed to deliver the therapeutic and imaging ligands into the tumor area, and outlined methods for enhancement of the tumor ablative ability of the NPS, including several important combinatorial approaches. In addition, we have summarized different in vitro and in vivo effects of PB NP-based therapies used to overcome both systemic and tumor-associated local barriers. An important new approach - PB NP-based immune drug delivery, which is an exciting and promising strategy to overcome cancer resistance and tumor recurrence - has been discussed. Finally, we have discussed the current understanding of the toxicological effects of PB NPs and PB NP-based therapeutics. We conclude that PB NP-based multimodal imaging-guided chemo-photothermal therapy offers new treatment strategies to overcome current hurdles in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Milan Gautam
- College of Pharmacy, Yeungnam University, 214-1 Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Kishwor Poudel
- College of Pharmacy, Yeungnam University, 214-1 Dae-Dong, Gyeongsan 712-749, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1 Dae-Dong, Gyeongsan 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1 Dae-Dong, Gyeongsan 712-749, Republic of Korea.
| |
Collapse
|
128
|
Ribeiro C, Borges J, Costa AMS, Gaspar VM, Bermudez VDZ, Mano JF. Preparation of Well-Dispersed Chitosan/Alginate Hollow Multilayered Microcapsules for Enhanced Cellular Internalization. Molecules 2018; 23:E625. [PMID: 29534439 PMCID: PMC6017778 DOI: 10.3390/molecules23030625] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 02/07/2023] Open
Abstract
Hollow multilayered capsules have shown massive potential for being used in the biomedical and biotechnology fields, in applications such as cellular internalization, intracellular trafficking, drug delivery, or tissue engineering. In particular, hollow microcapsules, developed by resorting to porous calcium carbonate sacrificial templates, natural-origin building blocks and the prominent Layer-by-Layer (LbL) technology, have attracted increasing attention owing to their key features. However, these microcapsules revealed a great tendency to aggregate, which represents a major hurdle when aiming for cellular internalization and intracellular therapeutics delivery. Herein, we report the preparation of well-dispersed polysaccharide-based hollow multilayered microcapsules by combining the LbL technique with an optimized purification process. Cationic chitosan (CHT) and anionic alginate (ALG) were chosen as the marine origin polysaccharides due to their biocompatibility and structural similarity to the extracellular matrices of living tissues. Moreover, the inexpensive and highly versatile LbL technology was used to fabricate core-shell microparticles and hollow multilayered microcapsules, with precise control over their composition and physicochemical properties, by repeating the alternate deposition of both materials. The microcapsules' synthesis procedure was optimized to extensively reduce their natural aggregation tendency, as shown by the morphological analysis monitored by advanced microscopy techniques. The well-dispersed microcapsules showed an enhanced uptake by fibroblasts, opening new perspectives for cellular internalization.
Collapse
Affiliation(s)
- Carla Ribeiro
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
- Department of Chemistry and CQ-VR, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal.
| | - João Borges
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Ana M S Costa
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Vítor M Gaspar
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Verónica de Zea Bermudez
- Department of Chemistry and CQ-VR, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|