1
|
Sun T, Liu Y, Wang K, Duan F, Lu L. Biotransformation of Tyrosol into a Novel Valuable α-Galactoside with Increased Solubility and Improved Anti-inflammatory Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37319317 DOI: 10.1021/acs.jafc.3c02529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Herein, tyrosol [2-(4-hydroxyphenyl) ethanol], which is rich in olive oil and red wine, was converted to a novel bioactive galactoside by enzymic glycosylation. The gene of α-galactosidase from Geobacillus stearothermophilus 23 was cloned and expressed in Escherichia coli as catalytically active inclusion bodies. The catalytically active inclusion bodies efficiently catalyzed the galactosylation of tyrosol using either melibiose or raffinose family oligosaccharides as glycosyl donors, resulting in a glycoside with 42.2 or 14.2% yields. The glycoside product was purified and identified as p-hydroxyphenethyl α-d-galactopyranoside by mass spectrometry and NMR analyses. The inclusion bodies can be recycled and reused for at least 10 batch reactions of galactoside synthesis. Moreover, the galactoside showed 11-fold increased water solubility and reduced cytotoxicity as compared to tyrosol. Also, it exhibited higher antioxidative and anti-inflammatory activities than tyrosol based on lipopolysaccharide-induced activated BV2 cells. These results provided important insights into the application of tyrosol derivatives in functional foods.
Collapse
Affiliation(s)
- Tong Sun
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yan Liu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ke Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Feiyu Duan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Lili Lu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| |
Collapse
|
2
|
Dinh CT, Vu HT, Phan QTH, Nguyen LP, Tran TQ, Van Tran D, Quy NN, Pham DTN, Nguyen DT. Synthesis of glycyrrhetinic acid-modified liposomes to deliver Murrayafoline A for treatment of hepatocellular carcinoma. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:72. [PMID: 36195780 PMCID: PMC9532286 DOI: 10.1007/s10856-022-06692-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Hepatocellular carcinoma is a common type of cancer associated with a high mortality rate. Among several bioactive compounds, Murrayafoline A (MuA) has been proved as a bio substance that exhibits great potentials in treating liver cancer. In order to overcome the high cytotoxicity and low solubility of MuA, a delivery system based on nanocarriers is necessary to deliver MuA towards the desired target. In the present study, 18β-glycyrrhetinic acid (GA), which is known as a ligand for liver targeting, was used to construct the cholesterol-poly (ethylene glycol)-glycyrrhetinic acid (GA-PEG-Chol) conjugate and liposome for MuA administration. The compound was then examined for therapeutic efficacy and safety in HUVEC and HepG2 cells in 2D and 3D cell cultures. Results have shown that MuA-loaded liposomes had IC50 value of 2 µM in HepG2 and had the cytosolic absorption of 8.83 ± 0.97 ng/105 cells, while The IC50 value of MuA-loaded liposomes in HUVEC cell lines was 15 µM and the the cytosolic absorption was recorded as 3.62 ± 0.61 cells. The drug test on the 3D cancer sphere platform of the HepG2 cancer sphere showed that MuA-loaded GA liposomes had the highest efficacy at a concentration of 100 µg/mL. In short, these results suggest that MuA-loaded GA liposomes have the potential for maintenance drug delivery and liver targeting.
Collapse
Affiliation(s)
- Cuc Thi Dinh
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi, 10000, Vietnam
| | - Ha Thi Vu
- Institute of Natural Products Chemistry, 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi, 10000, Vietnam
| | - Quynh Thi Huong Phan
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi, 10000, Vietnam
| | - Linh Phuong Nguyen
- Hanoi Medical University, 1 Ton That Tung St., Dong Da Dist., Hanoi, 10000, Vietnam
| | - Toan Quoc Tran
- Institute of Natural Products Chemistry, 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi, 10000, Vietnam
- Graduate University of Science and Technology, 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi, 10000, Vietnam
| | - Dung Van Tran
- VIET ANH VENTURE INVESTMENT J.S. COMPANY USA SANFORDPHARMA FACTORY, Hanoi, 10000, Vietnam
| | - Nguyen Ngoc Quy
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam.
| | - Duong Thanh Nguyen
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi, 10000, Vietnam.
- Graduate University of Science and Technology, 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi, 10000, Vietnam.
| |
Collapse
|
3
|
Liposomal Formulation for Oral Delivery of Cyclosporine A: Usefulness as a Semisolid-Dispersion System. Pharm Res 2022; 39:977-987. [PMID: 35501532 DOI: 10.1007/s11095-022-03276-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE This study aims to understand the process and mechanism of oral drug absorption from liposomes and to verify the usefulness of liposomal formulation for poorly soluble drugs. METHODS Cyclosporine A (CsA) was used as a model drug and entrapped into Dipalmitoylphosphatidylcholine (DPPC) and distearoylphosphatidylcholine (DSPC) liposomes. Molecular state of CsA in the liposomes was analyzed using powder X-ray diffraction (PXRD) and polarized light microscopy (PLM). Release profiles of CsA from liposomes were observed in fasted state simulated intestinal fluid (FaSSIF). Oral absorption of CsA from liposomal formulations were investigated in rats. RESULTS PXRD and PLM analyses suggested that CsA exists in the lipid layer of liposomes as a molecular dispersed state. Although both liposomes retained CsA stably in the simple buffer, DPPC liposomes quickly released CsA within 10 min in FaSSIF due to the interaction with bile acid. In contrast, effect of bile acid was negligible in DSPC, indicating a high resistivity to membrane perturbation. Oral bioavailability of CsA from liposomal formulations were almost comparable with that from a marketed product (Neoral). However, the absorption profiles were clearly different. CsA was absorbed quickly from DPPC liposomes and Neoral, while sustained absorption profile was observed from DSPC liposomes. Further study in which ritonavir was co-entrapped in the liposomes with CsA showed the higher efficacy of ritonavir to increase oral bioavailability of CsA. CONCLUSION Liposomes allows the appropriate formulation design for oral delivery of poorly soluble drugs, not only to increase the extent but also to control the rate of absorption.
Collapse
|
4
|
Lu S, Zhao P, Deng Y, Liu Y. Mechanistic Insights and Therapeutic Delivery through Micro/Nanobubble-Assisted Ultrasound. Pharmaceutics 2022; 14:pharmaceutics14030480. [PMID: 35335857 PMCID: PMC8954263 DOI: 10.3390/pharmaceutics14030480] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/12/2022] [Accepted: 02/19/2022] [Indexed: 02/05/2023] Open
Abstract
Ultrasound with low frequency (20–100 kHz) assisted drug delivery has been widely investigated as a non-invasive method to enhance the permeability and retention effect of drugs. The functional micro/nanobubble loaded with drugs could provide an unprecedented opportunity for targeted delivery. Then, ultrasound with higher intensity would locally burst bubbles and release agents, thus avoiding side effects associated with systemic administration. Furthermore, ultrasound-mediated destruction of micro/nanobubbles can effectively increase the permeability of vascular membranes and cell membranes, thereby not only increasing the distribution concentration of drugs in the interstitial space of target tissues but also promoting the penetration of drugs through cell membranes into the cytoplasm. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theragnostic tool. In this review, we first discuss the structure and generation of micro/nanobubbles. Second, ultrasound parameters and mechanisms of therapeutic delivery are discussed. Third, potential biomedical applications of micro/nanobubble-assisted ultrasound are summarized. Finally, we discuss the challenges and future directions of ultrasound combined with micro/nanobubbles.
Collapse
|
5
|
Mukai H, Ogawa K, Kato N, Kawakami S. Recent advances in lipid nanoparticles for delivery of nucleic acid, mRNA, and gene editing-based therapeutics. Drug Metab Pharmacokinet 2022; 44:100450. [PMID: 35381574 PMCID: PMC9363157 DOI: 10.1016/j.dmpk.2022.100450] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/26/2022]
Abstract
Lipid nanoparticles (LNPs) are becoming popular as a means of delivering therapeutics, including those based on nucleic acids and mRNA. The mRNA-based coronavirus disease 2019 vaccines are perfect examples to highlight the role played by drug delivery systems in advancing human health. The fundamentals of LNPs for the delivery of nucleic acid- and mRNA-based therapeutics, are well established. Thus, future research on LNPs will focus on addressing the following: expanding the scope of drug delivery to different constituents of the human body, expanding the number of diseases that can be targeted, and studying the change in the pharmacokinetics of LNPs under physiological and pathological conditions. This review article provides an overview of recent advances aimed at expanding the application of LNPs, focusing on the pharmacokinetics and advantages of LNPs. In addition, analytical techniques, library construction and screening, rational design, active targeting, and applicability to gene editing therapy have also been discussed.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan.
| |
Collapse
|
6
|
Abstract
We here describe the synthesis and biological evaluation of glycan shields for cell penetrating peptides. A new benzyl alkoxyamine connector was employed for the coupling of two saccharides units in the lateral side chain of individual amino acids in a peptide sequence. The oxyme bond formation with the corresponding glycan aldehydes allowed the preparation of highly glycosylated penetrating peptides with a minimal synthetic effort. Surprisingly, it was found that a four to six saccharide substitution did not decrease uptake efficiency in cells, whereas it significantly improved the toxicity profile of the penetrating peptide. In particular, glucose substitution was confirmed as an optimal glycan shield that showed an excellent in vitro uptake and intracellular localization as well as a superior in vivo biodistribution.
Collapse
Affiliation(s)
- Iván Gallego
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
7
|
Metelkina O, Huck B, O'Connor JS, Koch M, Manz A, Lehr CM, Titz A. Targeting extracellular lectins of Pseudomonas aeruginosa with glycomimetic liposomes. J Mater Chem B 2022; 10:537-548. [PMID: 34985094 DOI: 10.1039/d1tb02086b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The antimicrobial resistance crisis requires novel approaches for the therapy of infections especially with Gram-negative pathogens. Pseudomonas aeruginosa is defined as priority 1 pathogen by the WHO and thus of particular interest. Its drug resistance is primarily associated with biofilm formation and essential constituents of its extracellular biofilm matrix are the two lectins, LecA and LecB. Here, we report microbial lectin-specific targeted nanovehicles based on liposomes. LecA- and LecB-targeted phospholipids were synthesized and used for the preparation of liposomes. These liposomes with varying surface ligand density were then analyzed for their competitive and direct lectin binding activity. We have further developed a microfluidic device that allowed the optical detection of the targeting process to the bacterial lectins. Our data showed that the targeted liposomes are specifically binding to their respective lectin and remain firmly attached to surfaces containing these lectins. This synthetic and biophysical study provides the basis for future application in targeted antibiotic delivery to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Olga Metelkina
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany. .,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124 Braunschweig, Germany.,Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
| | - Benedikt Huck
- Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jonathan S O'Connor
- KIST Europe, 66123 Saarbrücken, Germany.,Department of Systems Engineering, Saarland University, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Andreas Manz
- KIST Europe, 66123 Saarbrücken, Germany.,Department of Systems Engineering, Saarland University, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany. .,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124 Braunschweig, Germany.,Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
8
|
Esmail S, Manolson MF. Advances in understanding N-glycosylation structure, function, and regulation in health and disease. Eur J Cell Biol 2021; 100:151186. [PMID: 34839178 DOI: 10.1016/j.ejcb.2021.151186] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 01/17/2023] Open
Abstract
N-linked glycosylation is a post-translational modification crucial for membrane protein folding, stability and other cellular functions. Alteration of membrane protein N-glycans is implicated in wide range of pathological conditions including cancer metastasis, chronic inflammatory diseases, and viral pathogenesis. Even though the roles of N-glycans have been studied extensively, our knowledge of their mechanisms remains unclear due to the lack of detailed structural analysis of the N-glycome. Mapping the N-glycome landscape will open new avenues to explore disease mechanisms and identify novel therapeutic targets. This review discusses the diverse structure of N-linked glycans, the function and regulation of N-glycosylation in health and disease, and ends with a focus on recent approaches to target N-glycans in rheumatoid arthritis and cancer metastasis.
Collapse
Affiliation(s)
- Sally Esmail
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada.
| | - Morris F Manolson
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
9
|
Chia ZC, Yang LX, Cheng TY, Chen YJ, Cheng HL, Hsu FT, Wang YJ, Chen YY, Huang TC, Fang YS, Huang CC. In Situ Formation of Au-Glycopolymer Nanoparticles for Surface-Enhanced Raman Scattering-Based Biosensing and Single-Cell Immunity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:52295-52307. [PMID: 34706531 DOI: 10.1021/acsami.1c13647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Successful synthesis of glyconanoparticles has attracted much attention due to their various biointeractive capabilities, but it is still a challenge to understand different single-cell responses to exogenous particles among cell populations. Herein, we designed polyaniline-containing galactosylated gold nanoparticles (Au@PGlyco NPs) via in situ polymerization of ortho-nitrophenyl-β-galactoside assisted by Au nucleation. The nanogold-carrying polyaniline block produced electromagnetic enhancement in surface-enhanced Raman scattering (SERS). The underlying polymerization mechanism of ortho-nitrophenyl compounds via the formation of Au nanoparticles was investigated. Depending on how the galactoside moiety reacted with β-galactosidase derived from bacteria, the Au@PGlyco NPs-mediated SERS biosensor could detect low amounts of bacteria (∼1 × 102 CFU/mL). In addition, a high accumulation of Au@PGlyco NPs mediated the immune response of tumor-associated M2 macrophages to the immunogenic M1 macrophage transition, which was elicited by reactive oxygen levels biostimulation using single-cell SERS-combined fluorescence imaging. Our study suggested that Au@PGlyco NPs may serve as a biosensing platform with the labeling capacity on galactose-binding receptors expressed cell and immune regulation.
Collapse
Affiliation(s)
- Zi-Chun Chia
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Li-Xing Yang
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ting-Yu Cheng
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ya-Jyun Chen
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Horng-Long Cheng
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tzu-Chi Huang
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Syun Fang
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chih-Chia Huang
- Department of Photonics, National Cheng Kung University, Tainan 70101, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
10
|
Biglione C, Neumann‐Tran TMP, Kanwal S, Klinger D. Amphiphilic micro‐ and nanogels: Combining properties from internal hydrogel networks, solid particles, and micellar aggregates. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Catalina Biglione
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin Berlin Germany
| | | | - Sidra Kanwal
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin Berlin Germany
| | - Daniel Klinger
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin Berlin Germany
| |
Collapse
|
11
|
Liu H, Zhong W, Zhang X, Lin D, Wu J. Nanomedicine as a promising strategy for the theranostics of infectious diseases. J Mater Chem B 2021; 9:7878-7908. [PMID: 34611689 DOI: 10.1039/d1tb01316e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Infectious diseases caused by bacteria, viruses, and fungi and their global spread pose a great threat to human health. The 2019 World Health Organization report predicted that infection-related mortality will be similar to cancer mortality by 2050. Particularly, the global cumulative numbers of the recent outbreak of coronavirus disease (COVID-19) have reached 110.7 million cases and over 2.4 million deaths as of February 23, 2021. Moreover, the crisis of these infectious diseases exposes the many problems of traditional diagnosis, treatment, and prevention, such as time-consuming and unselective detection methods, the emergence of drug-resistant bacteria, serious side effects, and poor drug delivery. There is an urgent need for rapid and sensitive diagnosis as well as high efficacy and low toxicity treatments. The emergence of nanomedicine has provided a promising strategy to greatly enhance detection methods and drug treatment efficacy. Owing to their unique optical, magnetic, and electrical properties, nanoparticles (NPs) have great potential for the fast and selective detection of bacteria, viruses, and fungi. NPs exhibit remarkable antibacterial activity by releasing reactive oxygen species and metal ions, exerting photothermal effects, and causing destruction of the cell membrane. Nano-based delivery systems can further improve drug permeability, reduce the side effects of drugs, and prolong systemic circulation time and drug half-life. Moreover, effective drugs against COVID-19 are still lacking. Recently, nanomedicine has shown great potential to accelerate the development of safe and novel anti-COVID-19 drugs. This article reviews the fundamental mechanisms and the latest developments in the treatment and diagnosis of bacteria, viruses, and fungi and discusses the challenges and perspectives in the application of nanomedicine.
Collapse
Affiliation(s)
- Hengyu Liu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xinyu Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China. .,School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
12
|
Bioevaluation of glucose-modified liposomes as a potential drug delivery system for cancer treatment using 177-Lu radiotracking. J Control Release 2021; 332:301-311. [PMID: 33675880 DOI: 10.1016/j.jconrel.2021.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Abstract
Liposomes are promising drug's delivery systems due to decreased toxicity of the liposome-encapsulated drug, but wider clinical application requires their more efficient tumor targeting with uptake, controlled drug release and higher shelf life. The unique metabolic characteristics of cancer cells based on higher demand for energy and therefore increased glucose utilization were exploited in the design of glucose modified liposomes (GML) with the aim to provide increased tumor targeting via glucose transporters and increased ability of drug delivery into tumor cells. Tumor accumulating potential of GML and non-glucose liposomes (NGL) were investigated on CT26 and LS174T tumor-bearing mice by simple and reliable radiotracer method using 177Lu as radioactive marker. Both liposomes, GML and NGL were radiolabeled in high radiolabeling yield, showing high in vitro stability in biological media, as the main prerequisite for the biodistribution studies. Tumors displayed significantly better accumulation of 177Lu-GML with the maximum uptake 6 h post-injection (5.8 ± 0.2%/g in LS174T tumor and 5.1 ± 0.5%/g in CT26 tumor), compared to negligible uptake of 177Lu-NGL (0.6 ± 0.1%/g in LS174T tumor and 0.9 ± 0.2%/g in CT26 tumor). Results of comparative biodistribution studies of 177Lu-NGL and 177Lu-GML indicate that increased accumulation of GML is enabled by glucose transporters and subsequent endocytosis, resulting in their prolonged retention in tumor tissues (up to 72 h). Direct radiolabeling of liposomes with 177Lu may be used not only for biodistribution studies using radiotracking, but also for cancer treatment.
Collapse
|
13
|
Fumoto S, Nishida K. Co-delivery Systems of Multiple Drugs Using Nanotechnology for Future Cancer Therapy. Chem Pharm Bull (Tokyo) 2021; 68:603-612. [PMID: 32611997 DOI: 10.1248/cpb.c20-00008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer treatments have improved significantly during the last decade but are not yet satisfactory. Combination therapy is often administered to improve efficacy and safety. Drug delivery systems can also improve efficacy and safety. To control the spatiotemporal distribution of drugs, nanotechnology involving liposomes, solid lipid nanoparticles, and polymeric micelles has been developed. Co-delivery systems of multiple drugs are a promising approach to combat cancer. Synergistic effects and reduced side effects are expected from the use of co-delivery systems. In this review, we summarize various co-delivery systems for multiple drugs, including small-molecule drugs, nucleic acids, genes, and proteins. Co-delivery of drugs with different properties is relatively difficult, but some researchers have succeeded in developing such co-delivery systems. Environment-responsive carrier designs can control the release of cargos. Although their preparation is more complicated than that of mono-delivery systems, co-delivery systems can simplify clinical procedures and improve patient QOL.
Collapse
Affiliation(s)
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
14
|
Rivero-Barbarroja G, Benito JM, Ortiz Mellet C, García Fernández JM. Cyclodextrin-Based Functional Glyconanomaterials. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2517. [PMID: 33333914 PMCID: PMC7765426 DOI: 10.3390/nano10122517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/29/2022]
Abstract
Cyclodextrins (CDs) have long occupied a prominent position in most pharmaceutical laboratories as "off-the-shelve" tools to manipulate the pharmacokinetics of a broad range of active principles, due to their unique combination of biocompatibility and inclusion abilities. The development of precision chemical methods for their selective functionalization, in combination with "click" multiconjugation procedures, have further leveraged the nanoscaffold nature of these oligosaccharides, creating a direct link between the glyco and the nano worlds. CDs have greatly contributed to understand and exploit the interactions between multivalent glycodisplays and carbohydrate-binding proteins (lectins) and to improve the drug-loading and functional properties of nanomaterials through host-guest strategies. The whole range of capabilities can be enabled through self-assembly, template-assisted assembly or covalent connection of CD/glycan building blocks. This review discusses the advancements made in this field during the last decade and the amazing variety of functional glyconanomaterials empowered by the versatility of the CD component.
Collapse
Affiliation(s)
- Gonzalo Rivero-Barbarroja
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (G.R.-B.); (C.O.M.)
| | - Juan Manuel Benito
- Instituto de Investigaciones Químicas (IIQ), CSIC, Universidad de Sevilla, 41092 Sevilla, Spain;
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (G.R.-B.); (C.O.M.)
| | | |
Collapse
|
15
|
Yamashita S, Katsumi H, Shimizu E, Nakao Y, Yoshioka A, Fukui M, Kimura H, Sakane T, Yamamoto A. Dendrimer-based micelles with highly potent targeting to sites of active bone turnover for the treatment of bone metastasis. Eur J Pharm Biopharm 2020; 157:85-96. [PMID: 33039547 DOI: 10.1016/j.ejpb.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 01/29/2023]
Abstract
Bone-drug targeting therapies using nanoparticles based on targeting ligands remain challenging due to their uptake clearance at non-target sites such as the liver, kidney, and spleen. Furthermore, the distribution sites of nanoparticles in bones have not been fully investigated, thus halting the development of more effective bone metastasis treatment strategies. In this study, we developed nanoparticles self-assembled from cholesterol-terminated, polyethylene glycol-conjugated, aspartic acid (Asp)-modified polyamidoamine dendrimer (Asp-PAMAM-Micelles) with targeting to active bone turnover sites associated with bone metastasis pathogenesis. On analysis through whole-body single photon emission computed tomography/computed tomography (SPECT/CT) imaging, 111In-Asp-PAMAM-Micelles showed high specificity to active bone turnover sites (especially the joints in the lower limbs, shoulder, and pelvis) after intravenous injection in mice. The lower limb bone uptake clearance for 111In-Asp-PAMAM-Micelles encapsulating paclitaxel (PTX) was 3.5-fold higher than that for 111In-unmodified PAMAM-Micelles (PTX). 3H-PTX encapsulated Asp-PAMAM-Micelles effectively accumulated in the lower limb bones in a similar manner as the 111In-Asp-PAMAM-Micelles (PTX). In a bone metastatic tumor mouse model, the tumor growth in the lower limb bones was significantly inhibited by injection of Asp-PAMAM-Micelles (PTX) compared to unmodified PAMAM-Micelles (PTX). Our results demonstrate that Asp-PAMAM-Micelles are sophisticated drug delivery systems for highly potent targeting to active bone turnover sites.
Collapse
Affiliation(s)
- Shugo Yamashita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Erika Shimizu
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Yuto Nakao
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Ayane Yoshioka
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Minako Fukui
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan; Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
16
|
Katsumi H, Yamashita S, Morishita M, Yamamoto A. Bone-Targeted Drug Delivery Systems and Strategies for Treatment of Bone Metastasis. Chem Pharm Bull (Tokyo) 2020; 68:560-566. [DOI: 10.1248/cpb.c20-00017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Shugo Yamashita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| |
Collapse
|
17
|
Chen X, Hu X, Hu J, Qiu Z, Yuan M, Zheng G. Celastrol-Loaded Galactosylated Liposomes Effectively Inhibit AKT/c-Met-Triggered Rapid Hepatocarcinogenesis in Mice. Mol Pharm 2020; 17:738-747. [PMID: 31904241 DOI: 10.1021/acs.molpharmaceut.9b00428] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our previous study proved that celastrol was a potential candidate for hepatocellular carcinoma (HCC) therapy. However, poor water solubility and toxic side effects may restrict its clinical application. To overcome these shortcomings and optimize its antitumor efficacy, we developed galactosylated liposomes using galactose-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) to deliver celastrol (C-GPL). C-GPL improved the water solubility of celastrol and exhibited high encapsulation efficiency, good stability in serum, and slow drug release profile. In vitro studies showed that C-GPL increased the cellular uptake of celastrol through receptor-mediated endocytosis, thereby enhancing celastrol cytotoxicity and cancer cell apoptosis. Particularly, in vivo antitumor activity of C-GPL was assessed in rapid HCC mouse models established via hydrodynamic transfection of the activated forms of AKT and c-Met. Compared to free celastrol, C-GPL significantly prevented liver weight gain, decreased liver damage biomarkers (glutamic-oxalacetic transaminase and alanine aminotransferase) and HCC marker (alpha-fetoprotein), and led to tumor disappearance on the liver surface. The improved therapeutic effect of C-GPL may be attributed to suppression of AKT activation, induction of apoptosis, and retardation of cell proliferation. Importantly, C-GPL exerted low toxicity to normal tissues without causing severe weight loss in mice. Taken together, C-GPL may become a promising drug delivery system for HCC treatment.
Collapse
Affiliation(s)
- Xinyan Chen
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xianxian Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhenpeng Qiu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
18
|
Dalva M, Lavik IK, El Jellas K, Gravdal A, Lugea A, Pandol SJ, Njølstad PR, Waldron RT, Fjeld K, Johansson BB, Molven A. Pathogenic Carboxyl Ester Lipase (CEL) Variants Interact with the Normal CEL Protein in Pancreatic Cells. Cells 2020; 9:cells9010244. [PMID: 31963687 PMCID: PMC7017060 DOI: 10.3390/cells9010244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations in the gene encoding the digestive enzyme carboxyl ester lipase (CEL) are linked to pancreatic disease. The CEL variant denoted CEL-HYB predisposes to chronic pancreatitis, whereas the CEL-MODY variant causes MODY8, an inherited disorder of endocrine and exocrine pancreatic dysfunction. Both pathogenic variants exhibit altered biochemical and cellular properties compared with the normal CEL protein (CEL-WT, wild type). We here aimed to investigate effects of CEL variants on pancreatic acinar and ductal cell lines. Following extracellular exposure, CEL-HYB, CEL-MODY, and CEL-WT were endocytosed. The two pathogenic CEL proteins significantly reduced cell viability compared with CEL-WT. We also found evidence of CEL uptake in primary human pancreatic acinar cells and in native ductal tissue. Moreover, coexpression of CEL-HYB or CEL-MODY with CEL-WT affected secretion of the latter, as CEL-WT was observed to accumulate intracellularly to a higher degree in the presence of either pathogenic variant. Notably, in coendocytosis experiments, both pathogenic variants displayed a modest effect on cell viability when CEL-WT was present, indicating that the normal protein might diminish toxic effects conferred by CEL-HYB and CEL-MODY. Taken together, our findings provide valuable insight into how the pathogenic CEL variants predispose to pancreatic disease and why these disorders develop slowly over time.
Collapse
Affiliation(s)
- Monica Dalva
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Ida K. Lavik
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
| | - Khadija El Jellas
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Anny Gravdal
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Aurelia Lugea
- Pancreatic Research Group, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.L.); (S.J.P.); (R.T.W.)
| | - Stephen J. Pandol
- Pancreatic Research Group, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.L.); (S.J.P.); (R.T.W.)
| | - Pål R. Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Richard T. Waldron
- Pancreatic Research Group, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.L.); (S.J.P.); (R.T.W.)
| | - Karianne Fjeld
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Bente B. Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Correspondence: ; Tel.: +47-55971263
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway; (M.D.); (I.K.L.); (K.E.J.); (A.G.); (K.F.); (A.M.)
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway;
- Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| |
Collapse
|
19
|
Hashida M. Role of pharmacokinetic consideration for the development of drug delivery systems: A historical overview. Adv Drug Deliv Rev 2020; 157:71-82. [PMID: 32565225 DOI: 10.1016/j.addr.2020.06.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/13/2022]
Abstract
Drug delivery system is defined as a system or technology to achieve optimum therapeutic effects of drugs through precise control of their movements in the body. In order to optimize function of drug delivery systems aiming at targeting, their whole-body distribution profiles should be systematically evaluated and analyzed, where pharmacokinetic analysis based on the clearance concepts plays important role. Organ perfusion experiments combined with statistical moment analysis further supply detailed information on drug disposition at organ and cellular levels. Based on general relationship between physicochemical properties and distribution profile, macromolecular prodrugs or polymer conjugates of proteins are rationally designed and further introduction of ligand structure brings cell-specific delivery for them. These approaches are also applicable for particulate carriers such as liposomes and offer various opportunities for biological drugs such as nucleic acid drugs for their delivery. Mechanistic approach for dermal absorption analysis based on physiological skin model offers another opportunity in rational design of drug delivery. Potential of drug delivery technology in future medicines such as cell therapy and nanomaterial platform application is further discussed in relation to pharmacokinetic consideration.
Collapse
|
20
|
Chen F, Huang G. Application of glycosylation in targeted drug delivery. Eur J Med Chem 2019; 182:111612. [DOI: 10.1016/j.ejmech.2019.111612] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 01/10/2023]
|
21
|
Tunki L, Kulhari H, Vadithe LN, Kuncha M, Bhargava S, Pooja D, Sistla R. Modulating the site-specific oral delivery of sorafenib using sugar-grafted nanoparticles for hepatocellular carcinoma treatment. Eur J Pharm Sci 2019; 137:104978. [DOI: 10.1016/j.ejps.2019.104978] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/07/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022]
|
22
|
Gallego I, Rioboo A, Reina JJ, Díaz B, Canales Á, Cañada FJ, Guerra‐Varela J, Sánchez L, Montenegro J. Glycosylated Cell‐Penetrating Peptides (GCPPs). Chembiochem 2019; 20:1400-1409. [DOI: 10.1002/cbic.201800720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/22/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Iván Gallego
- Centro Singular de Investigación en Química Biolóxica e, Materiais Moleculares (CIQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela Campus Vida 15782 Santiago de Compostela Spain
| | - Alicia Rioboo
- Centro Singular de Investigación en Química Biolóxica e, Materiais Moleculares (CIQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela Campus Vida 15782 Santiago de Compostela Spain
| | - José J. Reina
- Centro Singular de Investigación en Química Biolóxica e, Materiais Moleculares (CIQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela Campus Vida 15782 Santiago de Compostela Spain
| | - Bernardo Díaz
- Centro de Investigaciones Biológicas (CIB) del CSIC C/Ramiro de Maetzu 9, CP 28040 Madrid Spain
- Departamento de Biología Estructural y QuímicaFac. Ciencias Químicas Univ. Complutense de Madrid Avd/ Complutense s/n, CP Madrid Spain
| | - Ángeles Canales
- Departamento de Biología Estructural y QuímicaFac. Ciencias Químicas Univ. Complutense de Madrid Avd/ Complutense s/n, CP Madrid Spain
| | - F. Javier Cañada
- Centro de Investigaciones Biológicas (CIB) del CSIC C/Ramiro de Maetzu 9, CP 28040 Madrid Spain
| | - Jorge Guerra‐Varela
- Departamento de Zooloxía, Xenética e Antropoloxía FísicaFacultade de Veterinaria Universidade de Santiago de Compostela 27002 Lugo Spain
| | - Laura Sánchez
- Departamento de Zooloxía, Xenética e Antropoloxía FísicaFacultade de Veterinaria Universidade de Santiago de Compostela 27002 Lugo Spain
| | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e, Materiais Moleculares (CIQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela Campus Vida 15782 Santiago de Compostela Spain
| |
Collapse
|
23
|
Synthesis of a high functionality and quality lipid with gp130 binding hydrophobic peptide for the preparation of human glioma cell-targeted PEGylated liposomes. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2018.12.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
24
|
Huang Y, Hu L, Huang S, Xu W, Wan J, Wang D, Zheng G, Xia Z. Curcumin-loaded galactosylated BSA nanoparticles as targeted drug delivery carriers inhibit hepatocellular carcinoma cell proliferation and migration. Int J Nanomedicine 2018; 13:8309-8323. [PMID: 30584302 PMCID: PMC6289229 DOI: 10.2147/ijn.s184379] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The main objective of this study was to develop novel BSA nanoparticles (BSA NPs) for improving the bioavailability of curcumin as an anticancer drug, and those BSA NPs were galactosylated for forming the curcumin-loaded galactosylated BSA nanoparticles (Gal-BSA-Cur NPs), thus enhancing their ability to target asialoglycoprotein receptor (ASGPR) overexpressed on hepatocellular carcinoma (HCC) cells. Materials and methods Gal-BSA-Cur NPs were prepared by the desolvation method and showed a spherical shape and well distribution with the average particle size of 116.24 nm. Results In vitro drug release assay exhibited that Gal-BSA-Cur NPs had higher release rates and improved the curcumin solubility. Cell uptake studies confirmed that Gal-BSA-Cur NPs could selectively recognize receptors on the surface of HCC (HepG2) cells and improve internalization ability of drug compared with BSA NPs-loaded curcumin (BSA-Cur NPs), which might be due to high affinity to galactose. Further, the effects of Gal-BSA-Cur NPs were evaluated by cytotoxicity assay, crystal violet assay, cell apoptosis assay, and wound healing assay, respectively, which revealed that Gal-BSA-Cur NPs could inhibit HepG2 cells proliferation, induce cell apoptosis, and inhibit cell migration. Conclusion Immunofluorescence staining has proved that the effects of Gal-BSA-Cur NPs related to the suppression of the nuclear factor κB-p65 (NF-κB-p65) expression in HepG2 cell nucleus. Therefore, these results indicate that novel Gal-BSA-Cur NPs are potential candidates for targeted curcumin delivery to HCC cells.
Collapse
Affiliation(s)
- Yike Huang
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing, China,
| | - Lu Hu
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Shan Huang
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing, China,
| | - Wanjun Xu
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing, China,
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Dandan Wang
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing, China,
| | - Guocan Zheng
- Analytical and Testing Center, Chongqing University, Chongqing, China
| | - Zhining Xia
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences and Innovative Drug Research Centre, Chongqing University, Chongqing, China,
| |
Collapse
|
25
|
Morsy MA, Nair AB. Prevention of rat liver fibrosis by selective targeting of hepatic stellate cells using hesperidin carriers. Int J Pharm 2018; 552:241-250. [DOI: 10.1016/j.ijpharm.2018.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/29/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022]
|
26
|
Bousseau S, Vergori L, Soleti R, Lenaers G, Martinez MC, Andriantsitohaina R. Glycosylation as new pharmacological strategies for diseases associated with excessive angiogenesis. Pharmacol Ther 2018; 191:92-122. [DOI: 10.1016/j.pharmthera.2018.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/01/2018] [Indexed: 02/07/2023]
|
27
|
Mvango S, Matshe WMR, Balogun AO, Pilcher LA, Balogun MO. Nanomedicines for Malaria Chemotherapy: Encapsulation vs. Polymer Therapeutics. Pharm Res 2018; 35:237. [PMID: 30324329 DOI: 10.1007/s11095-018-2517-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/03/2018] [Indexed: 12/29/2022]
Abstract
Malaria is one of the oldest infectious diseases that afflict humans and its history extends back for millennia. It was once prevalent throughout the globe but today it is mainly endemic to tropical regions like sub-Saharan Africa and South-east Asia. Ironically, treatment for malaria has existed for centuries yet it still exerts an enormous death toll. This contradiction is attributed in part to the rapid development of resistance by the malaria parasite to chemotherapeutic drugs. In turn, resistance has been fuelled by poor patient compliance to the relatively toxic antimalarial drugs. While drug toxicity and poor pharmacological potentials have been addressed or ameliorated with various nanomedicine drug delivery systems in diseases like cancer, no clinically significant success story has been reported for malaria. There have been several reviews on the application of nanomedicine technologies, especially drug encapsulation, to malaria treatment. Here we extend the scope of the collation of the nanomedicine research literature to polymer therapeutics technology. We first discuss the history of the disease and how a flurry of scientific breakthroughs in the latter part of the nineteenth century provided scientific understanding of the disease. This is followed by a review of the disease biology and the major antimalarial chemotherapy. The achievements of nanomedicine in cancer and other infectious diseases are discussed to draw parallels with malaria. A review of the current state of the research into malaria nanomedicines, both encapsulation and polymer therapeutics polymer-drug conjugation technologies, is covered and we conclude with a consideration of the opportunities and challenges offered by both technologies.
Collapse
Affiliation(s)
- Sindisiwe Mvango
- Biopolymer Modification & Therapeutics Lab, Polymers & Composites, Materials Science & Manufacturing, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria, 0001, South Africa.,Department of Chemistry, University of Pretoria, Pretoria, 0002, South Africa
| | - William M R Matshe
- Biopolymer Modification & Therapeutics Lab, Polymers & Composites, Materials Science & Manufacturing, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria, 0001, South Africa
| | - Abideen O Balogun
- Department of Medicine, Nottingham University Hospital, Nottingham, UK
| | - Lynne A Pilcher
- Department of Chemistry, University of Pretoria, Pretoria, 0002, South Africa
| | - Mohammed O Balogun
- Biopolymer Modification & Therapeutics Lab, Polymers & Composites, Materials Science & Manufacturing, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria, 0001, South Africa.
| |
Collapse
|
28
|
Liu X, Shao W, Zheng Y, Yao C, Peng L, Zhang D, Hu XY, Wang L. GSH-Responsive supramolecular nanoparticles constructed by β-d-galactose-modified pillar[5]arene and camptothecin prodrug for targeted anticancer drug delivery. Chem Commun (Camb) 2018; 53:8596-8599. [PMID: 28718478 DOI: 10.1039/c7cc04932c] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Supramolecular construction of a targeted and stimuli-responsive drug delivery system is still a challenging task. Herein, GSH-responsive supramolecular prodrug nanoparticles were constructed by the host-guest complexation between a β-d-galactose-functionalized water-soluble pillar[5]arene (GalP5) and a disulfide bond containing camptothecin prodrug (G). The obtained prodrug nanoparticles were stable under physiological conditions, whereas efficient drug release was triggered in a simulated tumor environment with high GSH concentration. In vitro studies revealed that these prodrug nanoparticles preferentially entered asialoglycoprotein receptor-overexpressing HepG2 cells due to the active targeting effect of galactose units. This active targeting effect resulted in the maximization of anticancer efficacy and reduction of the undesirable side effects to normal cells.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Wei Shao
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Yanjing Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Chenhao Yao
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Luming Peng
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Dongmei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Xiao-Yu Hu
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Leyong Wang
- Key Laboratory of Mesoscopic Chemistry of MOE and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China. and Institute for Natural & Synthetic Organic Chemistry and School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China
| |
Collapse
|
29
|
Hagimori M, Chinda Y, Suga T, Yamanami K, Kato N, Inamine T, Fuchigami Y, Kawakami S. Synthesis of high functionality and quality mannose-grafted lipids to produce macrophage-targeted liposomes. Eur J Pharm Sci 2018; 123:153-161. [PMID: 30030100 DOI: 10.1016/j.ejps.2018.07.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/09/2018] [Accepted: 07/17/2018] [Indexed: 01/12/2023]
Abstract
The mannose receptor, which is responsible for tumor invasion, proliferation, and metastasis in the tumor microenvironment, is overexpressed in tumor-associated macrophages. Mannose is commonly applied to PEGylated liposomes in macrophage-targeted cancer therapy. To develop a high functionality and quality (HFQ) lipid for macrophage-targeted liposomes, we designed a novel mannosylated lipid with improved mannose receptor binding affinity using serine-glycine repeats (SG)n. We synthesized Man(S)-(SG)5-SSK-K(Pal)2 using only a fluorenylmethyloxycarbonyl (Fmoc) protecting group solid-phase peptide synthesis method, which produced a high-quality lipid at a moderately good yield. We then prepared Man-(SG)5/PEGylated liposomes using a post-insertion technique to insert Man(S)-(SG)5-SSK-K(Pal)2 into the PEGylated liposomes. In vitro cell investigations revealed that the Man-(SG)5/PEGylated liposomes effectively associated with mouse peritoneal macrophages by interacting with the mannose receptors. The results suggest that we produced a novel high-quality, highly functional mannosylated lipid that is suitable for clinical drug delivery applications.
Collapse
Affiliation(s)
- Masayori Hagimori
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan.
| | - Yorinao Chinda
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tadaharu Suga
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Kazuto Yamanami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tatsuo Inamine
- Department of Pharmacotherapeutics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Yuki Fuchigami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan.
| |
Collapse
|
30
|
Huang KW, Lai YT, Chern GJ, Huang SF, Tsai CL, Sung YC, Chiang CC, Hwang PB, Ho TL, Huang RL, Shiue TY, Chen Y, Wang SK. Galactose Derivative-Modified Nanoparticles for Efficient siRNA Delivery to Hepatocellular Carcinoma. Biomacromolecules 2018; 19:2330-2339. [PMID: 29808997 DOI: 10.1021/acs.biomac.8b00358] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Successful siRNA therapy requires suitable delivery systems with targeting moieties such as small molecules, peptides, antibodies, or aptamers. Galactose (Gal) residues recognized by the asialoglycoprotein receptor (ASGPR) can serve as potent targeting moieties for hepatocellular carcinoma (HCC) cells. However, efficient targeting to HCC via galactose moieties rather than normal liver tissues in HCC patients remains a challenge. To achieve more efficient siRNA delivery in HCC, we synthesized various galactoside derivatives and investigated the siRNA delivery capability of nanoparticles modified with those galactoside derivatives. In this study, we assembled lipid/calcium/phosphate nanoparticles (LCP NPs) conjugated with eight types of galactoside derivatives and demonstrated that phenyl β-d-galactoside-decorated LCP NPs (L4-LCP NPs) exhibited a superior siRNA delivery into HCC cells compared to normal hepatocytes. VEGF siRNAs delivered by L4-LCP NPs downregulated VEGF expression in HCC in vitro and in vivo and led to a potent antiangiogenic effect in the tumor microenvironment of a murine orthotopic HCC model. The efficient delivery of VEGF siRNA by L4-LCP NPs that resulted in significant tumor regression indicates that phenyl galactoside could be a promising HCC-targeting ligand for therapeutic siRNA delivery to treat liver cancer.
Collapse
Affiliation(s)
- Kuan-Wei Huang
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Yu-Tsung Lai
- Department of Chemistry , National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Guann-Jen Chern
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Shao-Feng Huang
- Department of Chemistry , National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Chia-Lung Tsai
- Department of Chemistry , National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan.,Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Cheng-Chin Chiang
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Pi-Bei Hwang
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Ting-Lun Ho
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Rui-Lin Huang
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Ting-Yun Shiue
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu 30013 , Taiwan.,Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , Hsinchu 30013 , Taiwan
| | - Sheng-Kai Wang
- Department of Chemistry , National Tsing Hua University , Hsinchu 30013 , Taiwan.,Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , Hsinchu 30013 , Taiwan
| |
Collapse
|
31
|
Cai L, Gu Z, Zhong J, Wen D, Chen G, He L, Wu J, Gu Z. Advances in glycosylation-mediated cancer-targeted drug delivery. Drug Discov Today 2018; 23:1126-1138. [DOI: 10.1016/j.drudis.2018.02.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/22/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022]
|
32
|
Ogawa K, Fuchigami Y, Hagimori M, Fumoto S, Miura Y, Kawakami S. Efficient gene transfection to the brain with ultrasound irradiation in mice using stabilized bubble lipopolyplexes prepared by the surface charge regulation method. Int J Nanomedicine 2018; 13:2309-2320. [PMID: 29713163 PMCID: PMC5907898 DOI: 10.2147/ijn.s157375] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Introduction We previously developed anionic ternary bubble lipopolyplexes, an ultrasound-responsive carrier, expecting safe and efficient gene transfection. However, bubble lipopolyplexes have a low capacity for echo gas (C3F8) encapsulation (EGE) in nonionic solution such as 5% glucose. On the other hand, we were able to prepare bubble lipopolyplexes by inserting phosphate-buffered saline before C3F8 encapsulation. Surface charge regulation (SCR) by electrolytes stabilizes liposome/plasmid DNA (pDNA) complexes by accelerated membrane fusion. Considering these facts, we hypothesized that SCR by electrolytes such as NaCl would promote C3F8 encapsulation in bubble lipopolyplexes mediated by accelerated membrane fusion. We defined this hypothesis as SCR-based EGE (SCR-EGE). Bubble lipopolyplexes prepared by the SCR-EGE method (SCR-EGE bubble lipopolyplexes) are expected to facilitate the gene transfection because of the high amount of C3F8. Therefore, we applied these methods for gene delivery to the brain and evaluated the characteristics of transgene expression in the brain. Methods First, we measured the encapsulation efficiency of C3F8 in SCR-EGE bubble lipopolyplexes. Next, we applied these bubble lipopolyplexes to the mouse brain; then, we evaluated the transfection efficiency. Furthermore, three-dimensional transgene distribution was observed using multicolor deep imaging. Results SCR-EGE bubble lipopolyplexes had a higher C3F8 content than conventional bubble lipopolyplexes. In terms of safety, SCR-EGE bubble lipopolyplexes possessed an anionic potential and showed no aggregation with erythrocytes. After applying SCR-EGE bubble lipopolyplexes to the brain, high transgene expression was observed by combining with ultrasound irradiation. As a result, transgene expression mediated by SCR-EGE bubble lipopolyplexes was observed mainly on blood vessels and partially outside of blood vessels. Conclusion The SCR-EGE method may promote C3F8 encapsulation in bubble lipopolyplexes, and SCR-EGE bubble lipopolyplexes may be potent carriers for efficient and safe gene transfection in the brain, especially to the blood vessels.
Collapse
Affiliation(s)
- Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuki Fuchigami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masayori Hagimori
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shintaro Fumoto
- Department of Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yusuke Miura
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
33
|
Bojarová P, Křen V. Sugared biomaterial binding lectins: achievements and perspectives. Biomater Sci 2018; 4:1142-60. [PMID: 27075026 DOI: 10.1039/c6bm00088f] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lectins, a distinct group of glycan-binding proteins, play a prominent role in the immune system ranging from pathogen recognition and tuning of inflammation to cell adhesion or cellular signalling. The possibilities of their detailed study expanded along with the rapid development of biomaterials in the last decade. The immense knowledge of all aspects of glycan-lectin interactions both in vitro and in vivo may be efficiently used in bioimaging, targeted drug delivery, diagnostic and analytic biological methods. Practically applicable examples comprise photoluminescence and optical biosensors, ingenious three-dimensional carbohydrate microarrays for high-throughput screening, matrices for magnetic resonance imaging, targeted hyperthermal treatment of cancer tissues, selective inhibitors of bacterial toxins and pathogen-recognising lectin receptors, and many others. This review aims to present an up-to-date systematic overview of glycan-decorated biomaterials promising for interactions with lectins, especially those applicable in biology, biotechnology or medicine. The lectins of interest include galectin-1, -3 and -7 participating in tumour progression, bacterial lectins from Pseudomonas aeruginosa (PA-IL), E. coli (Fim-H) and Clostridium botulinum (HA33) or DC-SIGN, receptors of macrophages and dendritic cells. The spectrum of lectin-binding biomaterials covered herein ranges from glycosylated organic structures, calixarene and fullerene cores over glycopeptides and glycoproteins, functionalised carbohydrate scaffolds of cyclodextrin or chitin to self-assembling glycopolymer clusters, gels, micelles and liposomes. Glyconanoparticles, glycan arrays, and other biomaterials with a solid core are described in detail, including inorganic matrices like hydroxyapatite or stainless steel for bioimplants.
Collapse
Affiliation(s)
- P Bojarová
- Laboratory of Biotransformation, Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, CZ 14220 Prague 4, Czech Republic.
| | - V Křen
- Laboratory of Biotransformation, Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, CZ 14220 Prague 4, Czech Republic.
| |
Collapse
|
34
|
Interaction of antitumoral drug erlotinib with biodegradable triblock copolymers: a molecular modeling study. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0413-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
35
|
Chen J, Chen Y, Cheng Y, Gao Y, Zheng P, Li C, Tong Y, Li Z, Luo W, Chen Z. Modifying glycyrrhetinic acid liposomes with liver-targeting ligand of galactosylated derivative: preparation and evaluations. Oncotarget 2017; 8:102046-102066. [PMID: 29254224 PMCID: PMC5731934 DOI: 10.18632/oncotarget.22143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
In this study, novel glycyrrhetinic acid (GA) liposomes modified with a liver-targeting galactosylated derivative ligand (Gal) were prepared using a film-dispersion method. To characterize the samples, particle size, zeta potential, drug loading, and encapsulation efficiency were performed. Moreover, plasma and tissues were pre-treated by liquid-liquid extraction and analyzed by high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). The results showed that the mean residence times (MRTs) and the area under the curve (AUC) of GA liposomes with Gal (Gal-GA-LP), and GA liposomes (GA-LP) were higher than the GA solution (GA-S) in plasma. The tissue (liver) distribution of Gal-GA-LP was significantly different in contrast to GA-LP. The relative intake rate (Re) of Gal-GA-LP and GA-LP in the liver was 4.752 and 2.196, respectively. The peak concentration ratio (Ce) of Gal-GA-LP and GA-LP in the liver was 2.796 and 1.083, respectively. The targeting efficiency (Te) of Gal-GA-LP and GA-LP in the liver was 48.193% and 34.718%, respectively. Taken together, the results indicate that Gal-GA-LP is an ideal complex for liver-targeting, and has great potential application in the clinical treatment of hepatic diseases. Drug loading and releasing experiments also indicated that most liposomes are spherical structures and have good dispersity under physiologic conditions, which could prolong GA release efficiency in vitro.
Collapse
Affiliation(s)
- Jing Chen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yuchao Chen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yi Cheng
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Youheng Gao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Pinjing Zheng
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Chuangnan Li
- The Second School of Clinic Medicine, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yidan Tong
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Zhao Li
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Wenhui Luo
- Guangdong Second Traditional Chinese Medicine Hospital (Guangdong Research Institute of Traditional Chinese Medicine Engineering Technology), Guangdong, China
| | - Zhao Chen
- Guangdong Second Traditional Chinese Medicine Hospital (Guangdong Research Institute of Traditional Chinese Medicine Engineering Technology), Guangdong, China
| |
Collapse
|
36
|
Chen J, Ding J, Xu W, Sun T, Xiao H, Zhuang X, Chen X. Receptor and Microenvironment Dual-Recognizable Nanogel for Targeted Chemotherapy of Highly Metastatic Malignancy. NANO LETTERS 2017. [PMID: 28644032 DOI: 10.1021/acs.nanolett.7b02129] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Targeted delivery of chemotherapeutic drugs to the desired lesion sites is the main objective in malignancy treatment, especially in highly metastatic malignancies. However, extensive studies around the world on traditional targeting strategies of recognizing either overexpressed receptors or microenvironments in tumors show great limitations, owing to the off-target effect and tumor homogeneity. Integration of both receptor-mediated targeting (RMT) and environment-mediated targeting (EMT) enhances the tumor accumulation and subsequent cell uptake at the same time, which may avoid these limitations. Herein, a dual targeting nanogel of PMNG engineered with both phenylboronic acid (PBA) and morpholine (MP) was reported for not only RMT via specific recognition of sialyl (SA) epitopes but also EMT toward extracellular acidity. Further engineering the nanoparticles via loading doxorubicin (DOX) brought a novel dual targeting system, that is, PMNG/DOX. PMNG/DOX demonstrated a greater targeting effect to both primary and metastatic B16F10 melanoma than the single PBA-modified nanogel (PNG) with only RMT in vitro and in vivo. Moreover, PMNG/DOX was also proved to be highly potent on inhibiting primary tumor growth as well as tumor metastasis on B16F10 melanoma-grafted mouse model. The results demonstrated the dual targeting design as a translational approach for drug delivery to highly metastatic tumor.
Collapse
Affiliation(s)
- Jinjin Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
- University of Chinese Academy of Sciences , Beijing 100039, People's Republic of China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Tianmeng Sun
- The First Hospital and Institute of Immunology, Jilin University , Changchun 130061, People's Republic of China
| | - Haihua Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| |
Collapse
|
37
|
Zaidi S, Misba L, Khan AU. Nano-therapeutics: A revolution in infection control in post antibiotic era. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2281-2301. [PMID: 28673854 DOI: 10.1016/j.nano.2017.06.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/20/2017] [Accepted: 06/20/2017] [Indexed: 12/22/2022]
Abstract
With the arrival of antibiotics 70 years ago, meant a paradigm shift in overcoming infectious diseases. For decades, drugs have been used to treat different infections. However, with time bacteria have become resistant to multiple antibiotics, making some diseases difficult to fight. Nanoparticles (NPs) as antibacterial agents appear to have potential to overcome such problems and to revolutionize the diagnosis and treatment of bacterial infections. Therefore, there is significant interest in the use of NPs to treat variety of infections, particularly caused by multidrug-resistant (MDR) strains. This review begins with illustration of types of NPs followed by the literature of current research addressing mechanisms of NPs antibacterial activity, steps involved in NP mediated drug delivery as well as areas where NPs use has potential to improve the treatment, like NP enabled vaccination. Besides, recently emerged innovative NP platforms have been highlighted and their progress made in each area has been reviewed.
Collapse
Affiliation(s)
- Sahar Zaidi
- Medical Microbiology and Molecular Biology Lab., Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Lama Misba
- Medical Microbiology and Molecular Biology Lab., Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Asad U Khan
- Medical Microbiology and Molecular Biology Lab., Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India.
| |
Collapse
|
38
|
Studzian M, Szulc A, Janaszewska A, Appelhans D, Pułaski Ł, Klajnert-Maculewicz B. Mechanisms of Internalization of Maltose-Modified Poly(propyleneimine) Glycodendrimers into Leukemic Cell Lines. Biomacromolecules 2017; 18:1509-1520. [DOI: 10.1021/acs.biomac.7b00046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | - Dietmar Appelhans
- Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069 Dresden, Germany
| | - Łukasz Pułaski
- Laboratory
of Transcriptional Regulation, Institute of Medical Biology PAS, Lodowa 106, 93-232 Lodz, Poland
| | | |
Collapse
|
39
|
Thao LQ, Lee C, Kim B, Lee S, Kim TH, Kim JO, Lee ES, Oh KT, Choi HG, Yoo SD, Youn YS. Doxorubicin and paclitaxel co-bound lactosylated albumin nanoparticles having targetability to hepatocellular carcinoma. Colloids Surf B Biointerfaces 2017; 152:183-191. [DOI: 10.1016/j.colsurfb.2017.01.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 12/22/2022]
|
40
|
Nanoformulation of dual bexarotene-tailed phospholipid conjugate with high drug loading. Eur J Pharm Sci 2017; 100:197-204. [DOI: 10.1016/j.ejps.2017.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/03/2017] [Accepted: 01/11/2017] [Indexed: 11/22/2022]
|
41
|
Liu X, Han M, Xu J, Geng S, Zhang Y, Ye X, Gou J, Yin T, He H, Tang X. Asialoglycoprotein receptor-targeted liposomes loaded with a norcantharimide derivative for hepatocyte-selective targeting. Int J Pharm 2017; 520:98-110. [DOI: 10.1016/j.ijpharm.2017.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
|
42
|
Staegemann MH, Gitter B, Dernedde J, Kuehne C, Haag R, Wiehe A. Mannose-Functionalized Hyperbranched Polyglycerol Loaded with Zinc Porphyrin: Investigation of the Multivalency Effect in Antibacterial Photodynamic Therapy. Chemistry 2017; 23:3918-3930. [PMID: 28029199 DOI: 10.1002/chem.201605236] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 02/03/2023]
Abstract
The antibacterial photodynamic activity of hyperbranched polyglycerol (hPG) loaded with zinc porphyrin photosensitizers and mannose units was investigated. hPG, with a MW of 19.5 kDa, was functionalized with about 15 molecules of the photosensitizer {5,10,15-tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) by using copper(I)-catalyzed 1,3-dipolar cycloaddition (CuAAC). These nanoparticle conjugates were functionalized systematically with increasing loadings of mannose in the range of approximately 20 to 110 groups. With higher mannose loadings (ca. 58-110 groups) the water-insoluble zinc porphyrin photosensitizer could thus be transferred into a water-soluble form. Targeting of the conjugates was proven in binding studies to the mannose-specific lectin concanavalin A (Con A) by using surface plasmon resonance (SPR). The antibacterial phototoxicity of the conjugates on Staphylococcus aureus (as a typical Gram-positive germ) was investigated in phosphate-buffered saline (PBS). It was shown that conjugates with approximately 70-110 mannose units exhibit significant antibacterial activity, whereas conjugates with approximately 20-60 units did not induce bacterial killing at all. These results give an insight into the multivalency effect in combination with photodynamic therapy (PDT). On addition of serum to the bacterial cultures, a quenching of this antibacterial phototoxicity was observed. In fluorescence studies with the conjugates in the presence of increasing bovine serum albumin (BSA) concentrations, protein-conjugate associations could be identified as a plausible cause for this quenching.
Collapse
Affiliation(s)
- Michael H Staegemann
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Burkhard Gitter
- Biolitec research GmbH, Otto-Schott-Str. 15, 07745, Jena, Germany
| | - Jens Dernedde
- Charité-Universitätsmedizin Berlin, Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christian Kuehne
- Charité-Universitätsmedizin Berlin, Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Arno Wiehe
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany.,Biolitec research GmbH, Otto-Schott-Str. 15, 07745, Jena, Germany
| |
Collapse
|
43
|
Li YH, Guo M, Shi SW, Zhang QL, Yang SP, Liu JG. A ruthenium-nitrosyl-functionalized nanoplatform for the targeting of liver cancer cells and NIR-light-controlled delivery of nitric oxide combined with photothermal therapy. J Mater Chem B 2017; 5:7831-7838. [DOI: 10.1039/c7tb02059g] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A multifunctional nanoplatform is capable of targeting liver cancer cells for NIR-light-controlled NO-release, and achieving both photodynamic and photothermal therapies.
Collapse
Affiliation(s)
- Yan-Hui Li
- Key Lab for Advanced Materials
- School of Chemistry & Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Min Guo
- Key Lab for Advanced Materials
- School of Chemistry & Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Shu-Wen Shi
- Key Lab for Advanced Materials
- School of Chemistry & Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Qian-Ling Zhang
- Shenzhen Key Lab of Functional Polymer
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- P. R. China
| | - Shi-Ping Yang
- Key Lab of Resource Chemistry of MOE & Shanghai Key Lab of Rare Earth Functional Materials
- Shanghai Normal University
- Shanghai
- P. R. China
| | - Jin-Gang Liu
- Key Lab for Advanced Materials
- School of Chemistry & Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| |
Collapse
|
44
|
Shi G, Du Y, Li Y, An Y, He Z, Lin Y, Zhang R, Yan X, Zhao J, Yang S, Brendan PNK, Liu F. Cell Recognition Molecule L1 Regulates Cell Surface Glycosylation to Modulate Cell Survival and Migration. Int J Med Sci 2017; 14:1276-1283. [PMID: 29104485 PMCID: PMC5666562 DOI: 10.7150/ijms.20479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 09/12/2017] [Indexed: 01/14/2023] Open
Abstract
Background: Cell recognition molecule L1 (L1) plays an important role in cancer cell differentiation, proliferation, migration and survival, but its mechanism remains unclear. Methodology/Principal: Our previous study has demonstrated that L1 enhanced cell survival and migration in neural cells by regulating cell surface glycosylation. In the present study, we show that L1 affected cell migration and survival in CHO (Chinese hamster ovary) cell line by modulation of sialylation and fucosylation at the cell surface via the PI3K (phosphoinositide 3-kinase) and Erk (extracellularsignal-regulated kinase) signaling pathways. Flow cytometry analysis indicated that L1 modulated cell surface sialylation and fucosylation in CHO cells. Activated L1 upregulated the protein expressions of ST6Gal1 (β-galactoside α-2,6-sialyltransferase 1) and FUT9 (Fucosyltransferase 9) in CHO cells. Furthermore, activated L1 promoted CHO cells migration and survival as shown by transwell assay and MTT assay. Inhibitors of sialylation and fucosylation blocked L1-induced cell migration and survival, while decreasing FUT9 and ST6Gal1 expressions via the PI3K-dependent and Erk-dependent signaling pathways. Conclusion : L1 modulated cell migration and survival by regulation of cell surface sialylation and fucosylation via the PI3K-dependent and Erk-dependent signaling pathways.
Collapse
Affiliation(s)
- Gang Shi
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | - Yue Du
- Dalian Medical University, Dalian, Liaoning 116044, China
| | - Yali Li
- National University Hospital, Singapore 119074, Singapore
| | - Yue An
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, China
| | - Zhenwei He
- Department of Neurology, Forth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Yingwei Lin
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, China
| | - Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | - Xiaofei Yan
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | - Jianfeng Zhao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | - Shihua Yang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | | | - Fang Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| |
Collapse
|
45
|
Coxon TP, Fallows TW, Gough JE, Webb SJ. A versatile approach towards multivalent saccharide displays on magnetic nanoparticles and phospholipid vesicles. Org Biomol Chem 2016; 13:10751-61. [PMID: 26360423 DOI: 10.1039/c5ob01591j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A simple synthetic route has been devised for the production of coating agents that can give multivalent displays of saccharides on the surface of magnetite nanoparticles and phospholipid vesicles. A versatile and potentially high-throughput condensation reaction allowed the rapid synthesis of a variety of glycosylhydrazide conjugates with lipid, resorcinol or catechol termini, each in good yield and high anomeric purity. The hydrolytic stability of these adducts was assessed in D2O at different pD values using (1)H-NMR spectroscopy, whilst quartz crystal microbalance with dissipation monitoring (QCM-D) confirmed that the saccharide functionality on bilayers and on nanoparticles was still available to lectins. These multivalent saccharide displays promoted nanoparticle interactions with cells, for example N-acetylglucosamine-coated nanoparticles interacted much more effectively with 3T3 fibroblasts than uncoated nanoparticles with these cells. Despite potential sensitivity to oxidation, catechol coatings on magnetite nanoparticles were found to be more stable and generate better nanoparticle interactions with fibroblasts than resorcinol coatings.
Collapse
Affiliation(s)
- Thomas P Coxon
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK. and School of Chemistry, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Thomas W Fallows
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK. and School of Chemistry, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Julie E Gough
- School of Materials, University of Manchester, MSS Tower, M13 9PL, Manchester, UK.
| | - Simon J Webb
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK. and School of Chemistry, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
46
|
Vieira AC, Chaves LL, Pinheiro M, Ferreira D, Sarmento B, Reis S. Design and statistical modeling of mannose-decorated dapsone-containing nanoparticles as a strategy of targeting intestinal M-cells. Int J Nanomedicine 2016; 11:2601-17. [PMID: 27354792 PMCID: PMC4907709 DOI: 10.2147/ijn.s104908] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of the present work was to develop and optimize surface-functionalized solid lipid nanoparticles (SLNs) for improvement of the therapeutic index of dapsone (DAP), with the application of a design of experiments. The formulation was designed to target intestinal microfold (M-cells) as a strategy to increase internalization of the drug by the infected macrophages. DAP-loaded SLNs and mannosylated SLNs (M-SLNs) were successfully developed by hot ultrasonication method employing a three-level, three-factor Box–Behnken design, after the preformulation study was carried out with different lipids. All the formulations were systematically characterized regarding their diameter, polydispersity index (PDI), zeta potential (ZP), entrapment efficiency, and loading capacity. They were also subjected to morphological studies using transmission electron microscopy, in vitro release study, infrared analysis (Fourier transform infrared spectroscopy), calorimetry studies (differential scanning calorimetry), and stability studies. The diameter of SLNs, SLN-DAP, M-SLNs, and M-SLN-DAP was approximately 300 nm and the obtained PDI was <0.2, confirming uniform populations. Entrapment efficiency and loading capacity were approximately 50% and 12%, respectively. Transmission electron microscopy showed spherical shape and nonaggregated nanoparticles. Fourier transform infrared spectroscopy was used to confirm the success of mannose coating process though Schiff’s base formation. The variation of the ZP between uncoated (approximately −30 mV) and mannosylated formulations (approximately +60 mV) also confirmed the successful coating process. A decrease in the enthalpy and broadening of the lipid melting peaks of the differential scanning calorimetry thermograms are consistent with the nanostructure of the SLNs. Moreover, the drug release was pH-sensitive, with a faster drug release at acidic pH than at neutral pH. Storage stability for the formulations for at least 8 weeks is expected, since they maintain the original characteristics of diameter, PDI, and ZP. These results pose a strong argument that the developed formulations can be explored as a promising carrier for treating leprosy with an innovative approach to target DAP directly to M-cells.
Collapse
Affiliation(s)
- Alexandre Cc Vieira
- UCIBIO, REQUIMTE, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Luíse L Chaves
- UCIBIO, REQUIMTE, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marina Pinheiro
- UCIBIO, REQUIMTE, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Domingos Ferreira
- Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Bruno Sarmento
- I3S, Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal; CESPU, Institute of Research and Advanced Formation in Health Sciences and Technology, University Institute of Health Sciences, Gandra, Portugal
| | - Salette Reis
- UCIBIO, REQUIMTE, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
47
|
Singh A, Ahmad I, Ahmad S, Iqbal Z, Ahmad FJ. A novel monolithic controlled delivery system of resveratrol for enhanced hepatoprotection: nanoformulation development, pharmacokinetics and pharmacodynamics. Drug Dev Ind Pharm 2016; 42:1524-36. [PMID: 26902951 DOI: 10.3109/03639045.2016.1151032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The current investigation aims to present a novel solid lipid-based nanoparticulate system of resveratrol (RV) for the effective treatment of liver cirrhosis. A simplified solvent injection method was employed and the Box-Behnken experimental design was applied for optimization to get a window particle size of 150-200 nm having maximum entrapment efficiency as well as % release. Optimized resveratrol solid lipid nanoparticles (RV-SLNs) (SR-1) of appropriate characteristics (particle size = 191.1 ± 10.44 nm; zeta potential= -13.56 ± 4.14 mV; entrapment efficiency = 75.23 ± 3.85%; maximum % release = 80.53 ± 3.99%) were produced. Differential scanning calorimetry and X-ray diffraction studies were carried out which collectively proved the reduced crystallinity and stability enhancing the effect of the SLNs. Improved drug stability was further established by the appreciable shelf-life of the formulation from International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH)-recommended accelerated stability studies. In vivo studies revealed nearly five-fold increase in the bioavailability of SR-1 (AUC0→∞=3411 ± 170.34 µg/ml/h) as compared to RV suspension (AUC0→∞=653.5 ± 30.10 µg/ml/h). Pharmacodynamic data exhibited a significant decrease in the serum biomarker enzymes (serum glutamic oxalo-acetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and alkaline phosphatase) after oral administration of RV-SLNs as compared to control and marketed (SILYBON(®)) formulations against paracetamol-induced liver cirrhosis. The effect of the treatment was confirmed by the histopathology of the liver microtome sections. Finally, reverse transcriptase-polymerase chain reaction studies were conducted on isolated liver mRNA from SR-1 treated animals and significant down-regulation of tissue inhibitor of metalloproteinases-1 and nuclear factor-kB was witnessed.
Collapse
Affiliation(s)
- Anjali Singh
- a Nanoformulation Research Lab , Jamia Hamdard , New Delhi , India ;,b Department of Pharmaceutical Medicine, Faculty of Pharmacy , Jamia Hamdard , New Delhi , India
| | - Iqbal Ahmad
- a Nanoformulation Research Lab , Jamia Hamdard , New Delhi , India
| | - Sayeed Ahmad
- c Bioactive Natural Product Lab , Jamia Hamdard , New Delhi , India
| | - Zeenat Iqbal
- a Nanoformulation Research Lab , Jamia Hamdard , New Delhi , India
| | - Farhan J Ahmad
- a Nanoformulation Research Lab , Jamia Hamdard , New Delhi , India
| |
Collapse
|
48
|
Fumoto S, Nishimura K, Nishida K, Kawakami S. Three-Dimensional Imaging of the Intracellular Fate of Plasmid DNA and Transgene Expression: ZsGreen1 and Tissue Clearing Method CUBIC Are an Optimal Combination for Multicolor Deep Imaging in Murine Tissues. PLoS One 2016; 11:e0148233. [PMID: 26824850 PMCID: PMC4732687 DOI: 10.1371/journal.pone.0148233] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 12/01/2015] [Indexed: 01/17/2023] Open
Abstract
Evaluation methods for determining the distribution of transgene expression in the body and the in vivo fate of viral and non-viral vectors are necessary for successful development of in vivo gene delivery systems. Here, we evaluated the spatial distribution of transgene expression using tissue clearing methods. After hydrodynamic injection of plasmid DNA into mice, whole tissues were subjected to tissue clearing. Tissue clearing followed by confocal laser scanning microscopy enabled evaluation of the three-dimensional distribution of transgene expression without preparation of tissue sections. Among the tested clearing methods (ClearT2, SeeDB, and CUBIC), CUBIC was the most suitable method for determining the spatial distribution of transgene expression in not only the liver but also other tissues such as the kidney and lung. In terms of the type of fluorescent protein, the observable depth for green fluorescent protein ZsGreen1 was slightly greater than that for red fluorescent protein tdTomato. We observed a depth of ~1.5 mm for the liver and 500 μm for other tissues without preparation of tissue sections. Furthermore, we succeeded in multicolor deep imaging of the intracellular fate of plasmid DNA in the murine liver. Thus, tissue clearing would be a powerful approach for determining the spatial distribution of plasmid DNA and transgene expression in various murine tissues.
Collapse
Affiliation(s)
- Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Koyo Nishimura
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shigeru Kawakami
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
49
|
Current applications of nanoparticles in infectious diseases. J Control Release 2016; 224:86-102. [PMID: 26772877 DOI: 10.1016/j.jconrel.2016.01.008] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 02/06/2023]
Abstract
For decades infections have been treated easily with drugs. However, in the 21st century, they may become lethal again owing to the development of antimicrobial resistance. Pathogens can become resistant by means of different mechanisms, such as increasing the time they spend in the intracellular environment, where drugs are unable to reach therapeutic levels. Moreover, drugs are also subject to certain problems that decrease their efficacy. This requires the use of high doses, and frequent administrations must be implemented, causing adverse side effects or toxicity. The use of nanoparticle systems can help to overcome such problems and increase drug efficacy. Accordingly, there is considerable current interest in their use as antimicrobial agents against different pathogens like bacteria, virus, fungi or parasites, multidrug-resistant strains and biofilms; as targeting vectors towards specific tissues; as vaccines and as theranostic systems. This review begins with an overview of the different types and characteristics of nanoparticles used to deliver drugs to the target, followed by a review of current research and clinical trials addressing the use of nanoparticles within the field of infectious diseases.
Collapse
|
50
|
Wang Z, Luo T, Sheng R, Li H, Sun J, Cao A. Amphiphilic Diblock Terpolymer PMAgala-b-P(MAA-co-MAChol)s with Attached Galactose and Cholesterol Grafts and Their Intracellular pH-Responsive Doxorubicin Delivery. Biomacromolecules 2015; 17:98-110. [DOI: 10.1021/acs.biomac.5b01227] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Zhao Wang
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Ting Luo
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Ruilong Sheng
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Hui Li
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Jingjing Sun
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Amin Cao
- CAS Key Laboratory of Synthetic
and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| |
Collapse
|