51
|
Liu Y, Chen Y, Lin L, Li H. Gambogic Acid as a Candidate for Cancer Therapy: A Review. Int J Nanomedicine 2020; 15:10385-10399. [PMID: 33376327 PMCID: PMC7764553 DOI: 10.2147/ijn.s277645] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Gambogic acid (GA), a kind of dry resin secreted by the Garcinia hanburyi tree, is a natural active ingredient with various biological activities, such as anti-cancer, anti-inflammatory, antioxidant, anti-bacterial effects, etc. An increasing amount of evidence indicates that GA has obvious anti-cancer effects via various molecular mechanisms, including the induction of apoptosis, autophagy, cell cycle arrest and the inhibition of invasion, metastasis, angiogenesis. In order to improve the efficacy in cancer treatment, nanometer drug delivery systems have been employed to load GA and form micelles, nanoparticles, nanofibers, and so on. In this review, we aim to offer a summary of chemical structure and properties, anti-cancer activities, drug delivery systems and combination therapy of GA, which might provide a reference to promote the development and clinical application of GA.
Collapse
Affiliation(s)
- Yuling Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yingchong Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
52
|
β-Cyclodextrin modified Pt(II) metallacycle-based supramolecular hyperbranched polymer assemblies for DOX delivery to liver cancer cells. Proc Natl Acad Sci U S A 2020; 117:30942-30948. [PMID: 33229542 DOI: 10.1073/pnas.2007798117] [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: 01/22/2023] Open
Abstract
Despite the widespread clinical application of chemotherapeutic anticancer drugs, their adverse side effects and inefficient performances remain ongoing issues. A drug delivery system (DDS) designed for a specific cancer may therefore overcome the drawbacks of single chemotherapeutic drugs and provide precise and synergistical cancer treatment by introducing exclusive stimulus responsiveness and combined chemotherapy properties. Herein, we report the design and synthesis of a supramolecular drug delivery assembly 1 constructed by orthogonal self-assembly technique in aqueous media specifically for application in liver cancer therapy. Complex 1 incorporates the β-cyclodextrin host molecule-functionalized organoplatinum(II) metallacycle 2 with two specific stimulus-responsive motifs to the signaling molecule nitric oxide (NO), in addition to the three-armed polyethylene glycol (PEG) functionalized ferrocene 3 with redox responsiveness. With this molecular design, the particularly low critical aggregation concentration (CAC) of assembly 1 allowed encapsulation of the commercial anticancer drug doxorubicin (DOX). Controlled drug release was also achieved by morphological transfer via a sensitive response to the endogenous redox and NO stimuli, which are specifically related to the microenvironment of liver tumor cells. Upon combination of these properties with the anticancer ability from the platinum acceptor, in vitro studies demonstrated that DOX-loaded 1 is able to codeliver anticancer drugs and exhibit therapeutic effectiveness to liver tumor sites via a synergistic effect, thereby revealing a potential DDS platform for precise liver cancer therapeutics.
Collapse
|
53
|
Wang C, Xia Y, Huo S, Shou D, Mei Q, Tang W, Li Y, Liu H, Zhou Y, Zhu B. Silencing of MEF2D by siRNA Loaded Selenium Nanoparticles for Ovarian Cancer Therapy. Int J Nanomedicine 2020; 15:9759-9770. [PMID: 33304100 PMCID: PMC7723231 DOI: 10.2147/ijn.s270441] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Delivery of therapeutic small interfering RNA (siRNA) via functionalized nanoparticles holds great promise for cancer therapy. However, developing a safe and efficient delivery carrier of siRNA is a challenging issue. METHODS RGDfC peptide was used to modify the surface of selenium nanoparticles (SeNPs) to synthesize a biocompatible siRNA delivery vehicle (R-SeNPs), and MEF2D-siRNA was loaded onto R-SeNPs to prepare a functionalized selenium nanoparticle R-Se@MEF2D-siRNA. The chemical properties of R-SeNPs were characterized, and the anticancer efficacy as well as related mechanisms of R-Se@MEF2D-siRNA were further explored. RESULTS R-Se@MEF2D-siRNA was significantly taken up by SKOV3 cells and could enter SKOV3 cells mainly in the clathrin-associated endocytosis way. The result of in vitro siRNA release demonstrated that R-Se@MEF2D-siRNA could release MEF2D-siRNA quicker in a microenvironment simulating a lysosomal environment in tumor cells compared to a normal physiological environment. The results of qRT-PCR assay proved that R-Se@MEF2D-siRNA could effectively silence the expression of the MEF2D gene in SKOV3 cells. R-Se@MEF2D-siRNA remarkably suppressed the proliferation of SKOV3 cells and further triggered its apoptosis. In addition, R-Se@MEF2D-siRNA had the capability to disrupt mitochondrial membrane potential (MMP) in SKOV3 cells and resulted in the overproduction of reactive oxygen species (ROS), indicating that mitochondrial dysfunction and ROS generation played an important role in the apoptosis of SKOV3 cells induced by R-Se@MEF2D-siRNA. In vivo, R-Se@MEF2D-siRNA also exhibited excellent antitumor activity mainly through decreasing tumor cells proliferation and triggering their apoptosis in tumor-bearing nude mice. CONCLUSION R-Se@MEF2D-siRNA provides an alternative strategy for ovarian cancer treatment in the clinic.
Collapse
Affiliation(s)
- Changbing Wang
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510120, People’s Republic of China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou510230, People’s Republic of China
| | - Yu Xia
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510120, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou510180, People’s Republic of China
| | - Shaochuan Huo
- Department of Orthopedics, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen518048, People’s Republic of China
- Shenzhen Research Institute of Guangzhou University of Chinese Medicine, Shenzhen518048, People’s Republic of China
| | - Diwen Shou
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou510180, People’s Republic of China
| | - Qing Mei
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou510180, People’s Republic of China
| | - Wenjuan Tang
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou510180, People’s Republic of China
| | - Yinghua Li
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510120, People’s Republic of China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510120, People’s Republic of China
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou510180, People’s Republic of China
| | - Bing Zhu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510120, People’s Republic of China
| |
Collapse
|
54
|
Florczak A, Deptuch T, Lewandowska A, Penderecka K, Kramer E, Marszalek A, Mackiewicz A, Dams-Kozlowska H. Functionalized silk spheres selectively and effectively deliver a cytotoxic drug to targeted cancer cells in vivo. J Nanobiotechnology 2020; 18:177. [PMID: 33261651 PMCID: PMC7709326 DOI: 10.1186/s12951-020-00734-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022] Open
Abstract
Background Chemotherapy is often a first-line therapeutic approach for the treatment of a wide variety of cancers. Targeted drug delivery systems (DDSs) can potentially resolve the problem of chemotherapeutic drug off-targeting effects. Herein, we examined in vivo models to determine the efficacy of Her2-targeting silk spheres (H2.1MS1) as DDSs for delivering doxorubicin (Dox) to Her2-positive and Her2-negative primary and metastatic mouse breast cancers. Results The specific accumulation of H2.1MS1 spheres was demonstrated at the site of Her2-positive cancer. Dox delivered only by functionalized H2.1MS1 particles selectively inhibited Her2-positive cancer growth in primary and metastatic models. Moreover, the significant effect of the Dox dose and the frequency of treatment administration on the therapeutic efficacy was indicated. Although the control MS1 spheres accumulated in the lungs in Her2-positive metastatic breast cancer, the Dox-loaded MS1 particles did not treat cancer. Histopathological examination revealed no systemic toxicity after multiple administrations and at increased doses of Dox-loaded silk spheres. Although the studies were performed in immunocompetent mice, the H2.1MS1 silk spheres efficiently delivered the drug, which exerted a therapeutic effect. Conclusion Our results indicated that functionalized silk spheres that enable cell-specific recognition, cellular internalization, and drug release represent an efficient strategy for cancer treatment in vivo.![]()
Collapse
Affiliation(s)
- Anna Florczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| | - Tomasz Deptuch
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Anna Lewandowska
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Karolina Penderecka
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Elzbieta Kramer
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Marszalek
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| |
Collapse
|
55
|
Wang X, Qiu Y, Wang M, Zhang C, Zhang T, Zhou H, Zhao W, Zhao W, Xia G, Shao R. Endocytosis and Organelle Targeting of Nanomedicines in Cancer Therapy. Int J Nanomedicine 2020; 15:9447-9467. [PMID: 33268987 PMCID: PMC7701161 DOI: 10.2147/ijn.s274289] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines (NMs) have played an increasing role in cancer therapy as carriers to efficiently deliver therapeutics into tumor cells. For this application, the uptake of NMs by tumor cells is usually a prerequisite to deliver the cargo to intracellular locations, which mainly relies on endocytosis. NMs can enter cells through a variety of endocytosis pathways. Different endocytosis pathways exhibit different intracellular trafficking routes and diverse subcellular localizations. Therefore, a comprehensive understanding of endocytosis mechanisms is necessary for increasing cellular entry efficiency and to trace the fate of NMs after internalization. This review focuses on endocytosis pathways of NMs in tumor cells, mainly including clathrin- and caveolae-mediated endocytosis pathways, involving effector molecules, expression difference of those molecules between normal and tumor cells, as well as the intracellular trafficking route of corresponding endocytosis vesicles. Then, the latest strategies for NMs to actively employ endocytosis are described, including improving tumor cellular uptake of NMs by receptor-mediated endocytosis, transporter-mediated endocytosis and enabling drug activity by changing intracellular routes. Finally, active targeting strategies towards intracellular organelles are also mentioned. This review will be helpful not only in explicating endocytosis and the trafficking process of NMs and elucidating anti-tumor mechanisms inside the cell but also in rendering new ideas for the design of highly efficacious and cancer-targeted NMs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mengyan Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Conghui Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tianshu Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Huimin Zhou
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenxia Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rongguang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
56
|
Barani M, Sabir F, Rahdar A, Arshad R, Kyzas GZ. Nanotreatment and Nanodiagnosis of Prostate Cancer: Recent Updates. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1696. [PMID: 32872181 PMCID: PMC7559844 DOI: 10.3390/nano10091696] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022]
Abstract
The fabrication and development of nanomaterials for the treatment of prostate cancer have gained significant appraisal in recent years. Advancements in synthesis of organic and inorganic nanomaterials with charge, particle size, specified geometry, ligand attachment etc have resulted in greater biocompatibility and active targeting at cancer site. Despite all of the advances made over the years in discovering drugs, methods, and new biomarkers for cancer of the prostate (PCa), PCa remains one of the most troubling cancers among people. Early on, effective diagnosis is an essential part of treating prostate cancer. Prostate-specific antigen (PSA) or serum prostate-specific antigen is the best serum marker widely accessible for diagnosis of PCa. Numerous efforts have been made over the past decade to design new biosensor-based strategies for biomolecules detection and PSA miniaturization biomarkers. The growing nanotechnology is expected to have a significant effect in the immediate future on scientific research and healthcare. Nanotechnology is thus predicted to find a way to solve one of the most and long-standing problem, "early cancer detection". For early diagnosis of PCa biomarkers, different nanoparticles with different approaches have been used. In this review, we provide a brief description of the latest achievements and advances in the use of nanoparticles for PCa biomarker diagnosis.
Collapse
Affiliation(s)
- Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 7616914111, Iran;
| | - Fakhara Sabir
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - George Z. Kyzas
- Department of Chemistry, International Hellenic University, 65404 Kavala, Greece
| |
Collapse
|
57
|
Lin C, Zhang LJ, Li B, Zhang F, Shen QR, Kong GQ, Wang XF, Cui SH, Dai R, Cao WQ, Zhang P. Selenium-Containing Protein From Selenium-Enriched Spirulina platensis Attenuates High Glucose-Induced Calcification of MOVAS Cells by Inhibiting ROS-Mediated DNA Damage and Regulating MAPK and PI3K/AKT Pathways. Front Physiol 2020; 11:791. [PMID: 32733280 PMCID: PMC7363841 DOI: 10.3389/fphys.2020.00791] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/15/2020] [Indexed: 01/29/2023] Open
Abstract
Hyperglycemia is the main feature of diabetes and may increase the risk of vascular calcification (VC), which is an independent predictor for cardiovascular and cerebrovascular diseases (CCD). Selenium (Se) may decrease the risk of CCD, and previous studies confirmed that Se-containing protein from Se-enriched Spirulina platensis (Se-SP) exhibited novel antioxidant potential. However, the effect of Se-SP against VC has been not investigated. Herein, the protective effect and underlying mechanism of Se-SP against high glucose-induced calcification in mouse aortic vascular smooth muscle cells (MOVAS) were explored. Inductively coupled plasma atomic emission spectroscopy (ICP-AES) results showed time-dependent uptake of Se-SP in MOVAS cells, which significantly inhibited high glucose-induced abnormal proliferation. Se-SP co-treatment also effectively attenuated high glucose-induced calcification of MOVAS cells, followed by decreased activity and expression of alkaline phosphatase (ALP). Further investigation revealed that Se-SP markedly prevented reactive oxygen species (ROS)-mediated DNA damage in glucose-treated MOVAS cells. ROS inhibition by glutathione (GSH) effectively inhibited high glucose-induced calcification, indicating that Se-SP could act as ROS inhibitor to inhibit high glucose-induced DNA damage and calcification. Moreover, Se-SP dramatically attenuated high glucose-induced dysfunction of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol-3-kinase/AKT (PI3K/AKT) pathways. Se-SP after Se addition achieved enhanced potential in inhibiting high glucose-induced calcification, which validated that Se-SP as a new Se species could be a highly effective treatment for human CCD.
Collapse
Affiliation(s)
- Cong Lin
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Jun Zhang
- Department of Neurology, People's Hospital of Linyi Affiliated to Qingdao University, Linyi, China
| | - Bo Li
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Feng Zhang
- Physical Examination Center, Taian City Central Hospital, Taian, China
| | - Qing-Rong Shen
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Guo-Qing Kong
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Xiao-Fan Wang
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Shou-Hong Cui
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Rong Dai
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Wen-Qiang Cao
- Department of Biotechnology, Zhuhai Hopegenes Medical and Phamaceutical Institute, Zhuhai, China
| | - Pu Zhang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, China
| |
Collapse
|
58
|
Understanding fundamentals of hepatocellular carcinoma to design next-generation chitosan nano-formulations: Beyond chemotherapy stride. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
59
|
Recent advances in the management of non-infectious posterior uveitis. Int Ophthalmol 2020; 40:3187-3207. [PMID: 32617804 DOI: 10.1007/s10792-020-01496-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To review the current regimens and novel therapeutic modalities in various stages of research and development for the management of non-infectious posterior uveitis (NIPU). METHODS We performed a thorough review of current literature using PubMed, Google Scholar and Clinicaltrials.gov to identify the published literature about the available therapeutics and novel drugs/therapies in different stages of clinical trials. RESULTS The current management regimen for non-infectious posterior uveitis includes corticosteroids, immunomodulatory therapies and anti-metabolites. However, NIPU requires long-term management for efficacious remission of the disease and to prevent disease relapse. Long-term safety issues associated with steroids have led to efforts to develop novel therapeutic agents including biological response modulators and immunosuppressants. The current therapeutic agents in various stages of development include calcineurin inhibitors, biologic response modifiers and a more a comprehensive modalities like ocular gene therapy as well as novel drug delivery mechanisms for higher bioavailability to the target tissues, with minimal systemic effects. CONCLUSION Novel efficacious therapeutic modalities under development will help overcome the challenges associated with the traditional therapeutic agents.
Collapse
|
60
|
Exhibiting the diagnostic face of selenium nanoparticles as a radio-platform for tumor imaging. Bioorg Chem 2020; 100:103910. [DOI: 10.1016/j.bioorg.2020.103910] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/28/2020] [Accepted: 05/02/2020] [Indexed: 12/23/2022]
|
61
|
Rajkumar K, Mvs S, Koganti S, Burgula S. Selenium Nanoparticles Synthesized Using Pseudomonas stutzeri (MH191156) Show Antiproliferative and Anti-angiogenic Activity Against Cervical Cancer Cells. Int J Nanomedicine 2020; 15:4523-4540. [PMID: 32606692 PMCID: PMC7320886 DOI: 10.2147/ijn.s247426] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/20/2020] [Indexed: 01/02/2023] Open
Abstract
Purpose Selenium nanoparticles (SeNP) have several applications in the field of biotechnology, including their use as anti-cancer drugs. The purpose of the present study is to analyze the efficacy of green synthesis on the preparation of SeNP and its effect on their anti-cancer properties. Methods A bacterial strain isolated from a freshwater source was shown to efficiently synthesize SeNP with potential therapeutic properties. The quality and stability of the NP were studied by scanning electron microscopy, X-ray diffraction, zeta-potential and FTIR analysis. A cost-effective medium formulation from biowaste having 6% banana peel extract enriched with 0.25 mM tryptophan was used to synthesize the NP. The NP after optimization was used to analyze their anti-tumor and anti-angiogenic activity. For this purpose, first, the cytotoxicity of the NP against cancer cells was analyzed by MTT assay and then chorioallantoic membrane assay was performed to assess anti-angiogenic activity. Further, cell migration assay and clonogenic inhibition assay were performed to test the anti-tumor properties of SeNP. To assess the cytotoxicity of SeNP on healthy RBC, hemolysis assay was performed. Results The strain identified as Pseudomonas stutzeri (MH191156) produced phenazine carboxylic acid, which aids the conversion of Se oxyanions to reduced NP state, resulting in particles in the size range of 75 nm to 200 nm with improved stability and quality of SeNP, as observed by zeta (ξ) potential of the particles which was found to be −46.2 mV. Cytotoxicity of the SeNP was observed even at low concentrations such as 5 µg/mL against cervical cancer cell line, ie, HeLa cells. Further, neovascularization was inhibited by upto 30 % in CAMs of eggs coinoculated with SeNp when compared with untreated controls, indicating significant anti-angiogenic activity of SeNP. The NP also inhibited the invasiveness of HeLa cells as observed by decreased cell migration and clonogenic proliferation. These observations indicate significant anti-tumor and anti-angiogenic activity of the SeNP in cervical cancer cells. Conclusion P. stutzeri (MH191156) is an efficient source of Se NP production with potential anti-angiogenic and anti-tumor properties, particularly against cervical cancer cells.
Collapse
Affiliation(s)
- Karthik Rajkumar
- Department of Microbiology, Osmania University, Tarnaka, Hyderabad 500007, India
| | - Sandhya Mvs
- Department of Microbiology, Osmania University, Tarnaka, Hyderabad 500007, India
| | - Siva Koganti
- Center for Advanced Research, Sri Venkateswara Institute of Medical Sciences, Tirupati 517507, India
| | - Sandeepta Burgula
- Department of Microbiology, Osmania University, Tarnaka, Hyderabad 500007, India
| |
Collapse
|
62
|
Xia Y, Tang G, Wang C, Zhong J, Chen Y, Hua L, Li Y, Liu H, Zhu B. Functionalized selenium nanoparticles for targeted siRNA delivery silence Derlin1 and promote antitumor efficacy against cervical cancer. Drug Deliv 2020; 27:15-25. [PMID: 31830840 PMCID: PMC6968560 DOI: 10.1080/10717544.2019.1667452] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Small interfering RNA (siRNA) exhibits great potential as a novel therapeutic option due to its highly sequence-specific ability to silence genes. However, efficient and safe delivery carriers are required for developing novel therapeutic paradigms. Thus, the successful development of efficient delivery platforms for siRNA is a crucial issue for the development of siRNA-based drugs in cancer treatments. In this study, biocompatible selenium nanoparticles (SeNPs) were loaded with RGDfC peptide to fabricate tumor-targeting gene delivery vehicle RGDfC-SeNPs. Subsequently, RGDfC-SeNPs were loaded with Derlin1-siRNA to fabricate RGDfC-Se@siRNA, which are functionalized selenium nanoparticles. RGDfC-Se@siRNA showed greater uptake in HeLa cervical cancer cells in comparison with that in human umbilical vein endothelial cells (HUVECs), verifying the RGDfC-mediated specific uptake of RGDfC-Se@siRNA. RGDfC-Se@siRNA was capable of entering HeLa cells via clathrin-associated endocytosis, and showed faster siRNA release in a cancer cell microenvironment in comparison with a normal physiological environment. qPCR and western blotting assays both indicated that RGDfC-Se@siRNA exhibited an obvious gene silencing efficacy in HeLa cells. RGDfC-Se@siRNA suppressed the invasion, migration and the proliferation of HeLa cells, and triggered HeLa cell apoptosis. Moreover, RGDfC-Se@siRNA induced the disruption of mitochondrial membrane potentials. Meanwhile, RGDfC-Se@siRNA enhanced the generation of reactive oxygen species (ROS) in HeLa cell, suggesting that mitochondrial dysfunction mediated by ROS might play a significant role in RGDfC-Se@siRNA-induced apoptosis. Interestingly, RGDfC-SeNPs@siRNA exhibited significant antitumor activity in a HeLa tumor-bearing mouse model. Additionally, RGDfC-SeNPs@siRNA is nontoxic to main organ of mouse. The above results indicate that RGDfC-Se@siRNA provides a promising potential for cervical cancer therapy.
Collapse
Affiliation(s)
- Yu Xia
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Guoyi Tang
- Department of Obstetrics Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Changbing Wang
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiayu Zhong
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liang Hua
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yinghua Li
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Bing Zhu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
63
|
Shetab Boushehri MA, Dietrich D, Lamprecht A. Nanotechnology as a Platform for the Development of Injectable Parenteral Formulations: A Comprehensive Review of the Know-Hows and State of the Art. Pharmaceutics 2020; 12:pharmaceutics12060510. [PMID: 32503171 PMCID: PMC7356945 DOI: 10.3390/pharmaceutics12060510] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Within recent decades, the development of nanotechnology has made a significant contribution to the progress of various fields of study, including the domains of medical and pharmaceutical sciences. A substantially transformed arena within the context of the latter is the development and production of various injectable parenteral formulations. Indeed, recent decades have witnessed a rapid growth of the marketed and pipeline nanotechnology-based injectable products, which is a testimony to the remarkability of the aforementioned contribution. Adjunct to the ability of nanomaterials to deliver the incorporated payloads to many different targets of interest, nanotechnology has substantially assisted to the development of many further facets of the art. Such contributions include the enhancement of the drug solubility, development of long-acting locally and systemically injectable formulations, tuning the onset of the drug’s release through the endowment of sensitivity to various internal or external stimuli, as well as adjuvancy and immune activation, which is a desirable component for injectable vaccines and immunotherapeutic formulations. The current work seeks to provide a comprehensive review of all the abovementioned contributions, along with the most recent advances made within each domain. Furthermore, recent developments within the domains of passive and active targeting will be briefly debated.
Collapse
Affiliation(s)
- Maryam A. Shetab Boushehri
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-736428; Fax: +49-228-735268
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic of Bonn, 53105 Bonn, Germany;
| | - Alf Lamprecht
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- PEPITE EA4267, Institute of Pharmacy, University Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
64
|
Korany M, Mahmoud B, Ayoub SM, Sakr TM, Ahmed SA. Synthesis and radiolabeling of vitamin C-stabilized selenium nanoparticles as a promising approach in diagnosis of solid tumors. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07195-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
65
|
Zhou H, Gong Y, Liu Y, Huang A, Zhu X, Liu J, Yuan G, Zhang L, Wei JA, Liu J. Intelligently thermoresponsive flower-like hollow nano-ruthenium system for sustained release of nerve growth factor to inhibit hyperphosphorylation of tau and neuronal damage for the treatment of Alzheimer's disease. Biomaterials 2020; 237:119822. [DOI: 10.1016/j.biomaterials.2020.119822] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/26/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
|
66
|
Zhou T, Zhang W, Cheng D, Tang X, Feng J, Wu W. Preparation, Characterization, and in vivo Evaluation of NK4-Conjugated Hydroxycamptothecin-Loaded Liposomes. Int J Nanomedicine 2020; 15:2277-2286. [PMID: 32280220 PMCID: PMC7127779 DOI: 10.2147/ijn.s243746] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE In this study, NK4-conjugated hydroxycamptothecin liposomes (NK4-HCPT-Lips) were prepared with the aim of improving drug targeting to the liver. METHODS NK4-HCPT-Lips were prepared using the thin-film dispersion method. In vitro antitumor activities were evaluated by MTT assay. HCPT levels in plasma and tissues were determined via high-performance liquid chromatography (HPLC) with camptothecin as the internal standard, and the characteristics, pharmacokinetics, and bio-distribution of NK4-HCPT-Lips were evaluated. RESULTS The liposomes showed a regular spherical-shaped morphology, and the entrapment efficiency and drug loading capacity reached 82.5 ± 2.4% and 3.01 ± 0.23%, respectively, with a particle size of 155.6 ± 2.6 nm and a zeta potential of -24.8 ± 3.3 mV. Inhibition effect experiments found that NK4-HCPT-Lips had a good inhibition on the HepG2 cells. Pharmacokinetic studies revealed an increase in the area under the curve and mean residence time as well as a decrease in plasma clearance (p < 0.05) of the NK4-HCPT-Lips compared to those of HCPT liposomes and a commercial HCPT injection. Tissue distribution studies showed that NK4-HCPT-Lips were present at high levels in the liver but were cleared from the kidneys. CONCLUSION These results demonstrate that NK4-HCPT-Lips possess excellent liver-targeting attributes, which could enhance the therapeutic effects of drug treatments for hepatic diseases.
Collapse
Affiliation(s)
- Ting Zhou
- School of Pharmacy, Guilin Medical University, Guilin541004, People’s Republic of China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin541004, People’s Republic of China
| | - Dongliang Cheng
- School of Pharmacy, Guilin Medical University, Guilin541004, People’s Republic of China
| | - Xin Tang
- School of Public Health, Guilin Medical University, Guilin541004, People’s Republic of China
| | - Jianfang Feng
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning530200, People’s Republic of China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin541004, People’s Republic of China
| |
Collapse
|
67
|
Xia Y, Tang G, Guo M, Xu T, Chen H, Lin Z, Li Y, Chen Y, Zhu B, Liu H, Cao J. Silencing KLK12 expression via RGDfC-decorated selenium nanoparticles for the treatment of colorectal cancer in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110594. [PMID: 32204058 DOI: 10.1016/j.msec.2019.110594] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 02/07/2023]
Abstract
Short interfering RNA (siRNA) has been investigated as a promising modality of cancer treatment due to its capability to target specific target genes for downregulation. However, the successful application of this strategy depends on producing a safe and effective carrier system for delivering siRNA to the tumor. Thus, investigation of siRNA delivery carriers is a fundamental step in the field of siRNA-based therapeutics. In the current research, the surface of selenium nanoparticles (SeNPs) were modified with the tumor-targeted molecular RGDfC peptide with positive charge to synthetize the biocompatible siRNA carrier RGDfC-SeNPs. Subsequently, KLK12-siRNA was loaded onto the surface of RGDfC-SeNPs to create functionalized nanoparticles (RGDfC-Se@siRNA) that we tested for in vitro and in vivo antitumor efficacy. We measured significantly greater particle uptake in HT-29 colorectal cancer cells relative to HUVECs, providing evidence for the targeted delivery of RGDfC-Se@siRNA. We found that RGDfC-Se@siRNA could enter HT-29 cells primarily via clathrin-mediated endocytosis. Further, these particles experienced faster siRNA release in an acidic microenvironment compared to pH 7.4. The results from quantitative PCR and Western blot assays suggested that the target gene of KLK12 in HT-29 cells were obviously silenced by RGDfC-Se@siRNA. The further biological studies showed that treatment with RGDfC-Se@siRNA had ability to suppress the proliferation and migration/invasion of HT-29 cells, and triggered HT-29 cells apoptosis. RGDfC-Se@siRNA could induce the mitochondrial membrane potential (MMP) disruption and enhance the reactive oxygen species (ROS) generation in HT-29 cells, indicating that RGDfC-Se@siRNA induced the HT-29 cells apoptosis possibly by a ROS-mediated mitochondrial dysfunction pathway. Importantly, the in vivo antitumor study also verified that RGDfC-Se@siRNA could significantly suppress the growth of tumor in vivo. In addition, we did not observe any signs of systemic or tissue-specific toxicity after administration of RGDfC-Se@siRNA in mice. As a whole, these findings suggest that RGDfC-Se@siRNA has promising potential as a therapy for colorectal cancer.
Collapse
Affiliation(s)
- Yu Xia
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China.
| | - Guoyi Tang
- Department of Obstetrics Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Min Guo
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Tiantian Xu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Haiyang Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Zhengfang Lin
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yinghua Li
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yi Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Bing Zhu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| |
Collapse
|
68
|
Liu Y, Wei C, Lin A, Pan J, Chen X, Zhu X, Gong Y, Yuan G, Chen L, Liu J, Luo Z. Responsive functionalized MoSe 2 nanosystem for highly efficient synergistic therapy of breast cancer. Colloids Surf B Biointerfaces 2020; 189:110820. [PMID: 32045843 DOI: 10.1016/j.colsurfb.2020.110820] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/12/2020] [Accepted: 01/21/2020] [Indexed: 12/25/2022]
Abstract
The photothermal/photodynamic synergistic therapy is a promising tumor treatment, but developing nanosystems that achieve synchronous photothermal/photodynamic functions is still quite challenging. Here, we use a simple method to synthesize molybdenum selenide nanoparticles (MoSe2 NPs) with a photothermal effect as a carrier, and load a photosensitizer ICG to form a nanosystem (MoSe2@ICG-PDA-HA)with dual photothermal/photodynamic functions under near-infrared irradiation. In addition, the surface modification of the nanosystem with acid-responsive release polydopamine (PDA) and tumor-targeted hyaluronic acid (HA) enhanced the stability of the photosensitizer ICG and the accumulation of ICG at tumor sites. The multicellular sphere assay simulated solid tumors and demonstrated that MoSe2@ICG-PDA-HA could significantly inhibit the 4T1 cell growth. The anti-tumor experiments in tumor-bearing mice showed that MoSe2@ICG-PDA-HA not only significantly inhibited the growth of 4T1 subcutaneous tumors, but also inhibited their metastasis. This study presented a nanosystem that could improve the photostability of optical materials and enhance the photothermal/photodynamic synergy effect, providing a new idea for finding a way to effectively treat breast cancer.
Collapse
Affiliation(s)
- Yanan Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy,Guangdong Medical University, Zhanjiang, 524023, China; College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China; Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Chunfang Wei
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy,Guangdong Medical University, Zhanjiang, 524023, China; College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China; Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Ange Lin
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Jiali Pan
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Xu Chen
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Xufeng Zhu
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Youcong Gong
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Guanglong Yuan
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Lanmei Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy,Guangdong Medical University, Zhanjiang, 524023, China.
| | - Jie Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy,Guangdong Medical University, Zhanjiang, 524023, China; College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China; Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Zhaohui Luo
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China.
| |
Collapse
|
69
|
Zhong J, Xia Y, Hua L, Liu X, Xiao M, Xu T, Zhu B, Cao H. Functionalized selenium nanoparticles enhance the anti-EV71 activity of oseltamivir in human astrocytoma cell model. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3485-3491. [PMID: 31422717 DOI: 10.1080/21691401.2019.1640716] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Enterovirus 71 (EV71) which commonly caused the hand-foot-mouth disease (HFMD) has become one of public health challenges worldwide. However, no effective vaccines or drugs for this disease has been developed. Thus, there is an urgent need to find a new strategy for treating the EV71 infection. Oseltamivir (OT) is an effective antiviral agent, but continuous use of oseltamivir leads to a diminished therapeutic effect in the clinic. In order to improve the antiviral activity of oseltamivir, oseltamivir was loaded onto surfaces of selenium nanoparticles (SeNPs) to fabricate a functionalized antiviral nanoparticles SeNPs@OT. The size of SeNPs@OT was tested by TEM and dynamic light scattering. The chemical structure and elemental composition of SeNPs@OT were analyzed by FT-IR and EDX, respectively. SeNPs@OT exhibited good stability and effective drug release in serum and PBS. SeNPs@OT efficiently entered into human astrocyte U251 cells (host cells) via clathrin-associated endocytosis and inhibited EV71 proliferation, which could protect EV71-infected U251 cells from apoptosis through mitochondrial pathway. Furthermore, SeNPs@OT inhibited EV71 activity probably by reducing the generation of reactive oxygen species in EV71-infected U251 cells. Interestingly, SeNPs obviously enhanced antiviral activity of oseltamivir in the anti-EV71 cell model. Taken together, SeNPs@OT is a promising antiviral drug candidate for EV71 infection.
Collapse
Affiliation(s)
- Jiayu Zhong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University , Guangzhou , People's Republic of China.,Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Yu Xia
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Liang Hua
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Xiaomin Liu
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Misi Xiao
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Tiantian Xu
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Bing Zhu
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou 510120 , People's Republic of China
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University , Guangzhou , People's Republic of China
| |
Collapse
|
70
|
Długosz O, Szostak K, Staroń A, Pulit-Prociak J, Banach M. Methods for Reducing the Toxicity of Metal and Metal Oxide NPs as Biomedicine. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E279. [PMID: 31936311 PMCID: PMC7013649 DOI: 10.3390/ma13020279] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 01/10/2023]
Abstract
The rapid development of medicine has forced equally rapid progress in the field of pharmaceuticals. In connection with the expensive and time-consuming process of finding new drugs, great emphasis is put on the design and use of metal and metal oxides nanoparticles in nanomedicine. The main focus is on comprehensive presentation of both physicochemical properties and the possibilities of using, in particular, silver (Ag) and gold (Au) nanoparticles, as well as zinc oxide (ZnO) and titanium oxide (TiO2) nanoparticles as drug carriers and in the treatment of cancer. An important element of this subject is the possibility of occurrence of toxic effects of these nanoparticles. For this reason, possible mechanisms of toxic actions are presented, as well as methods used to reduce their toxicity to ensure the safety of drug carriers based on these nanostructures.
Collapse
Affiliation(s)
| | | | | | | | - Marcin Banach
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska St., 31-155 Cracow, Poland; (O.D.); (K.S.); (A.S.); (J.P.-P.)
| |
Collapse
|
71
|
Nie H, Qiu B, Yang QX, Zhao Y, Liu XM, Zhang YT, Liao FL, Zhang SY. Effect of gal/GalNAc regioisomerism in galactosylated liposomes on asialoglycoprotein receptor-mediated hepatocyte-selective targeting in vivo. J Liposome Res 2019; 31:79-89. [PMID: 31691619 DOI: 10.1080/08982104.2019.1682606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, we describe a novel synthesis of galactosylated lipids by lipase catalysis. Lactitol (Lac), galactose (Gal), or N-acetyl galactosamine (GalNAc) was coupled with cholesterol (CHS) as target head groups by enzyme-catalyzed regioselective esterification to produce three kinds of lipids: CHS-1-Gal, CHS-6-Gal, or CHS-6-GalNAc1. The biological effects of galactosylated lipids carrying different constitutional isomers of the pendent sugar species were investigated. LP-1-Gal (liposomes containing 5.0 molar% of CHS-1-Gal) showed strong binding to tetrameric lectins of Ricinus communis agglutinin (RCA120) in vitro, while LP-6-Gal (liposomes containing 5.0 molar% of CHS-6-Gal) and LP-6-GalNAc (liposomes containing 5.0 molar% of CHS-6-GalNAc) did not. After intravenous injection, LP-6-GalNAc, LP-1-Gal and LP-6-Gal rapidly disappeared from the blood and accumulated rapidly in liver (up to 74.88 ± 4.11%, 58.67 ± 5.75%, and 47.66 ± 4.56% of injected dose/g organ within 4 h, respectively). This is significantly higher than the uptake of unmodified liposomes (Unmod-LP) (18.67 ± 6.07%). Pre-injection of asialofetuin significantly inhibits liver uptake of Gal-liposomes (P < 0.01), with the degree of inhibition appearing in the following order: LP-6-GalNAc (73.29%) > LP-1-Gal (67.06%) > LP-6-Gal (53.61%). More importantly, LP-6-GalNAc was preferentially taken up by hepatocytes and the uptake ratio by parenchymal cells (PC) and nonparenchymal cells (NPC) (PC/NPC ratio) was 11.03 higher than LP-1-Gal (7.32), LP-6-Gal (5.83) and Unmod-LP (2.39). We suggest that liposomes containing the novel galactosylated lipid CHS-6-GalNAc have potential as drug delivery carriers for hepatocyte-selective targeting.
Collapse
Affiliation(s)
- Hua Nie
- Medical College of Jiaying University, Meizhou, China
| | - Bo Qiu
- Medical College of Jiaying University, Meizhou, China
| | - Qi-Xuan Yang
- Medical College of Jiaying University, Meizhou, China
| | - Ying Zhao
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou, China
| | - Xiao-Min Liu
- Meizhou Engineering Research Center for Hakka Health Care, Meizhou, China
| | - Ying-Ting Zhang
- Meizhou Engineering Research Center for Hakka Health Care, Meizhou, China
| | - Fu-Lin Liao
- School of Life Science, Jiaying University, Meizhou, China
| | | |
Collapse
|