1
|
Yu X, Zhang Y, Hou L, Qiao X, Zhang Y, Cheng H, Lu H, Chen J, Du L, Zheng Q, Hou J, Tong G. Increases in Cellular Immune Responses Due to Positive Effect of CVC1302-Induced Lysosomal Escape in Mice. Vaccines (Basel) 2023; 11:1718. [PMID: 38006050 PMCID: PMC10675172 DOI: 10.3390/vaccines11111718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
This study found a higher percentage of CD8+ T cells in piglets immunized with a CVC1302-adjuvanted inactivated foot-and-mouth disease virus (FMDV) vaccine. We wondered whether the CVC1302-adjuvanted inactivated FMDV vaccine promoted cellular immunity by promoting the antigen cross-presentation efficiency of ovalbumin (OVA) through dendritic cells (DCs), mainly via cytosolic pathways. This was demonstrated by the enhanced levels of lysosomal escape of OVA in the DCs loaded with OVA and CVC1302. The higher levels of ROS and significantly enhanced elevated lysosomal pH levels in the DCs facilitated the lysosomal escape of OVA. Significantly enhanced CTL activity levels was observed in the mice immunized with OVA-CVC1302. Overall, CVC1302 increased the cross-presentation of exogenous antigens and the cross-priming of CD8+ T cells by alkalizing the lysosomal pH and facilitating the lysosomal escape of antigens. These studies shed new light on the development of immunopotentiators to improve cellular immunity induced by vaccines.
Collapse
Affiliation(s)
- Xiaoming Yu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yuanyuan Zhang
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Liting Hou
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Xuwen Qiao
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yuanpeng Zhang
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Haiwei Cheng
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Haiyan Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Jin Chen
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Luping Du
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Qisheng Zheng
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Jibo Hou
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety—State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| |
Collapse
|
2
|
Chu JJ, Ji WB, Zhuang JH, Gong BF, Chen XH, Cheng WB, Liang WD, Li GR, Gao J, Yin Y. Nanoparticles-based anti-aging treatment of Alzheimer's disease. Drug Deliv 2022; 29:2100-2116. [PMID: 35850622 PMCID: PMC9302016 DOI: 10.1080/10717544.2022.2094501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Age is the strongest risk factor for Alzheimer's disease (AD). In recent years, the relationship between aging and AD has been widely studied, with anti-aging therapeutics as the treatment for AD being one of the mainstream research directions. Therapeutics targeting senescent cells have shown improvement in AD symptoms and cerebral pathological changes, suggesting that anti-aging strategies may be a promising alternative for AD treatment. Nanoparticles represent an excellent approach for efficiently crossing the blood-brain barrier (BBB) to achieve better curative function and fewer side effects. Thereby, nanoparticles-based anti-aging treatment may exert potent anti-AD therapeutic efficacy. This review discusses the relationship between aging and AD and the application and prospect of anti-aging strategies and nanoparticle-based therapeutics in treating AD.
Collapse
Affiliation(s)
- Jian-Jian Chu
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China.,Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wen-Bo Ji
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China.,Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian-Hua Zhuang
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Bao-Feng Gong
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Xiao-Han Chen
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Wen-Bin Cheng
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Wen-Danqi Liang
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Gen-Ru Li
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Targeted liposomal doxorubicin/ceramides combinations: the importance to assess the nature of drug interaction beyond bulk tumor cells. Eur J Pharm Biopharm 2022; 172:61-77. [PMID: 35104605 DOI: 10.1016/j.ejpb.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/26/2022]
Abstract
One of the major assets of anticancer nanomedicine is the ability to co-deliver drug combinations, as it enables targeting of different cellular populations and/or signaling pathways implicated in tumorigenesis and thus tackling tumor heterogeneity. Moreover, drug resistance can be circumvented, for example, upon co-encapsulation and delivery of doxorubicin and sphingolipids, as ceramides. Herein, the impact of short (C6) and long (C18) alkyl chain length ceramides on the nature of drug interaction, within the scope of combination with doxorubicin, was performed in bulk triple-negative breast cancer (TNBC) cells, as well as on the density of putative cancer stem cells and phenotype, including live single-cell tracking. C6- or C18-ceramide enabled a synergistic drug interaction in all conditions and (bulk) cell lines tested. However, differentiation among these two ceramides was reflected on the migratory potential of cancer cells, particularly significant against the highly motile MDA-MB-231 cells. This effect was supported by the downregulation of the PI3K/Akt pathway enabled by C6-ceramide and in contrast with C18-ceramide. The decrease of the migratory potential enabled by the targeted liposomal combinations is of high relevance in the context of TNBC, due to the underlying metastatic potential. Surprisingly, the nature of the drug interaction assessed at the level of bulk cancer cells revealed to be insufficient to predict whether a drug combination enables a decrease in the percentage of the master regulators of tumor relapse as ALDH+/high putative TNBC cancer stem cells, suggesting, for the first time, that it should be extended further down to this level.
Collapse
|
4
|
Companioni O, Mir C, Garcia-Mayea Y, LLeonart ME. Targeting Sphingolipids for Cancer Therapy. Front Oncol 2021; 11:745092. [PMID: 34737957 PMCID: PMC8560795 DOI: 10.3389/fonc.2021.745092] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids are an extensive class of lipids with different functions in the cell, ranging from proliferation to cell death. Sphingolipids are modified in multiple cancers and are responsible for tumor proliferation, progression, and metastasis. Several inhibitors or activators of sphingolipid signaling, such as fenretinide, safingol, ABC294640, ceramide nanoliposomes (CNLs), SKI-II, α-galactosylceramide, fingolimod, and sonepcizumab, have been described. The objective of this review was to analyze the results from preclinical and clinical trials of these drugs for the treatment of cancer. Sphingolipid-targeting drugs have been tested alone or in combination with chemotherapy, exhibiting antitumor activity alone and in synergism with chemotherapy in vitro and in vivo. As a consequence of treatments, the most frequent mechanism of cell death is apoptosis, followed by autophagy. Aslthough all these drugs have produced good results in preclinical studies of multiple cancers, the outcomes of clinical trials have not been similar. The most effective drugs are fenretinide and α-galactosylceramide (α-GalCer). In contrast, minor adverse effects restricted to a few subjects and hepatic toxicity have been observed in clinical trials of ABC294640 and safingol, respectively. In the case of CNLs, SKI-II, fingolimod and sonepcizumab there are some limitations and absence of enough clinical studies to demonstrate a benefit. The effectiveness or lack of a major therapeutic effect of sphingolipid modulation by some drugs as a cancer therapy and other aspects related to their mechanism of action are discussed in this review.
Collapse
Affiliation(s)
- Osmel Companioni
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Spanish Biomedical Research Network Center in Oncology, CIBERONC, Madrid, Spain
| |
Collapse
|
5
|
Acid ceramidase controls apoptosis and increases autophagy in human melanoma cells treated with doxorubicin. Sci Rep 2021; 11:11221. [PMID: 34045496 PMCID: PMC8159975 DOI: 10.1038/s41598-021-90219-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
Acid ceramidase (AC) is a lysosomal hydrolase encoded by the ASAH1 gene, which cleaves ceramides into sphingosine and fatty acid. AC is expressed at high levels in most human melanoma cell lines and may confer resistance against chemotherapeutic agents. One such agent, doxorubicin, was shown to increase ceramide levels in melanoma cells. Ceramides contribute to the regulation of autophagy and apoptosis. Here we investigated the impact of AC ablation via CRISPR-Cas9 gene editing on the response of A375 melanoma cells to doxorubicin. We found that doxorubicin activates the autophagic response in wild-type A375 cells, which effectively resist apoptotic cell death. In striking contrast, doxorubicin fails to stimulate autophagy in A375 AC-null cells, which rapidly undergo apoptosis when exposed to the drug. The present work highlights changes that affect melanoma cells during incubation with doxorubicin, in A375 melanoma cells lacking AC. We found that the remarkable reduction in recovery rate after doxorubicin treatment is strictly associated with the impairment of autophagy, that forces the AC-inhibited cells into apoptotic path.
Collapse
|
6
|
Fulfager AD, Yadav KS. Understanding the implications of co-delivering therapeutic agents in a nanocarrier to combat multidrug resistance (MDR) in breast cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102405] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
7
|
Adityan S, Tran M, Bhavsar C, Wu SY. Nano-therapeutics for modulating the tumour microenvironment: Design, development, and clinical translation. J Control Release 2020; 327:512-532. [DOI: 10.1016/j.jconrel.2020.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
|
8
|
Zhang Y, Chen J, Zhao Y, Weng L, Xu Y. Ceramide Pathway Regulators Predict Clinical Prognostic Risk and Affect the Tumor Immune Microenvironment in Lung Adenocarcinoma. Front Oncol 2020; 10:562574. [PMID: 33194633 PMCID: PMC7653182 DOI: 10.3389/fonc.2020.562574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/28/2020] [Indexed: 01/29/2023] Open
Abstract
Purpose The ceramide pathway is strongly associated with the regulation of tumor proliferation, differentiation, senescence, and apoptosis. This study aimed to explore the gene signatures, prognostic value, and immune-related effects of ceramide-regulated genes in lung adenocarcinoma (LUAD). Methods Public datasets of LUAD from The Cancer Genome Atlas and Gene Expression Omnibus were selected. Consensus clustering was adopted to classify LUAD patients, and a least absolute shrinkage and selection operator (LASSO) regression model was employed to develop a prognostic risk signature. CIBERSORT algorithm was used to estimate the association between the risk signature and the tumor immune microenvironment. Results Most of the 22 ceramide-regulated genes were differentially expressed between LUAD and normal samples. LUAD patients were classified into two subgroups (cluster 1 and 2) and cluster 2 was associated with a poor prognosis. Furthermore, a prognostic risk signature was developed based on the three ceramide-regulated genes, Cytochrome C (CYCS), V-rel reticuloendotheliosis viral oncogene homolog A (RELA) and Fas-associated via death domain (FADD). LUAD patients with low- and high-risk scores differed concerning the subtypes of tumor-infiltrating immune cells. A moderate to weak correlation was observed between the risk score and tumor-infiltrating immune cells. Conclusions Ceramide-regulated genes could predict clinical prognostic risk and affect the tumor immune microenvironment in LUAD.
Collapse
Affiliation(s)
- Yuan Zhang
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianbo Chen
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Fujian Medical University, Xiamen, China
| | - Yunan Zhao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lihong Weng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yiquan Xu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
Iv M, Ng NN, Nair S, Zhang Y, Lavezo J, Cheshier SH, Holdsworth SJ, Moseley ME, Rosenberg J, Grant GA, Yeom KW. Brain Iron Assessment after Ferumoxytol-enhanced MRI in Children and Young Adults with Arteriovenous Malformations: A Case-Control Study. Radiology 2020; 297:438-446. [PMID: 32930651 DOI: 10.1148/radiol.2020200378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Iron oxide nanoparticles are an alternative contrast agent for MRI. Gadolinium deposition has raised safety concerns, but it is unknown whether ferumoxytol administration also deposits in the brain. Purpose To investigate whether there are signal intensity changes in the brain at multiecho gradient imaging following ferumoxytol exposure in children and young adults. Materials and Methods This retrospective case-control study included children and young adults, matched for age and sex, with brain arteriovenous malformations who received at least one dose of ferumoxytol from January 2014 to January 2018. In participants who underwent at least two brain MRI examinations (subgroup), the first and last available examinations were analyzed. Regions of interests were placed around deep gray structures on quantitative susceptibility mapping and R2* images. Mean susceptibility and R2* values of regions of interests were recorded. Measurements were assessed by linear regression analyses: a between-group comparison of ferumoxytol-exposed and unexposed participants and a within-group (subgroup) comparison before and after exposure. Results Seventeen participants (mean age ± standard deviation, 13 years ± 5; nine male) were in the ferumoxytol-exposed (case) group, 21 (mean age, 14 years ± 5; 11 male) were in the control group, and nine (mean age, 12 years ± 6; four male) were in the subgroup. The mean number of ferumoxytol administrations was 2 ± 1 (range, one to four). Mean susceptibility (in parts per million [ppm]) and R2* (in inverse seconds [sec-1]) values of the dentate (case participants: 0.06 ppm ± 0.04 and 23.87 sec-1 ± 4.13; control participants: 0.02 ppm ± 0.03 and 21.7 sec-1 ± 5.26), substantia nigrae (case participants: 0.08 ppm ± 0.06 and 27.46 sec-1 ± 5.58; control participants: 0.04 ppm ± 0.05 and 24.96 sec-1 ± 5.3), globus pallidi (case participants: 0.14 ppm ± 0.05 and 30.75 sec-1 ± 5.14; control participants: 0.08 ppm ± 0.07 and 28.82 sec-1 ± 6.62), putamina (case participants: 0.03 ppm ± 0.02 and 20.63 sec-1 ± 2.44; control participants: 0.02 ppm ± 0.02 and 19.65 sec-1 ± 3.6), caudate (case participants: -0.1 ppm ± 0.04 and 18.21 sec-1 ± 3.1; control participants: -0.06 ppm ± 0.05 and 18.83 sec-1 ± 3.32), and thalami (case participants: 0 ppm ± 0.03 and 16.49 sec-1 ± 3.6; control participants: 0.02 ppm ± 0.02 and 18.38 sec-1 ± 2.09) did not differ between groups (susceptibility, P = .21; R2*, P = .24). For the subgroup, the mean interval between the first and last ferumoxytol administration was 14 months ± 8 (range, 1-25 months). Mean susceptibility and R2* values of the dentate (first MRI: 0.06 ppm ± 0.05 and 25.78 sec-1 ± 5.9; last MRI: 0.06 ppm ± 0.02 and 25.55 sec-1 ± 4.71), substantia nigrae (first MRI: 0.06 ppm ± 0.06 and 28.26 sec-1 ± 9.56; last MRI: 0.07 ppm ± 0.06 and 25.65 sec-1 ± 6.37), globus pallidi (first MRI: 0.13 ppm ± 0.07 and 27.53 sec-1 ± 8.88; last MRI: 0.14 ppm ± 0.06 and 29.78 sec-1 ± 6.54), putamina (first MRI: 0.03 ppm ± 0.03 and 19.78 sec-1 ± 3.51; last MRI: 0.03 ppm ± 0.02 and 19.73 sec-1 ± 3.01), caudate (first MRI: -0.09 ppm ± 0.05 and 21.38 sec-1 ± 4.72; last MRI: -0.1 ppm ± 0.05 and 18.75 sec-1 ± 2.68), and thalami (first MRI: 0.01 ppm ± 0.02 and 17.65 sec-1 ± 5.16; last MRI: 0 ppm ± 0.02 and 15.32 sec-1 ± 2.49) did not differ between the first and last MRI examinations (susceptibility, P = .95; R2*, P = .54). Conclusion No overall significant differences were found in susceptibility and R2* values of deep gray structures to suggest retained iron in the brain between ferumoxytol-exposed and unexposed children and young adults with arteriovenous malformations and in those exposed to ferumoxytol over time. © RSNA, 2020.
Collapse
Affiliation(s)
- Michael Iv
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Nathan N Ng
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Sid Nair
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Yi Zhang
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Jonathan Lavezo
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Samuel H Cheshier
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Samantha J Holdsworth
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Michael E Moseley
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Jarrett Rosenberg
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Gerald A Grant
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| | - Kristen W Yeom
- From the Department of Radiology, Division of Neuroimaging and Neurointervention (M.I.), Department of Pathology (J.L.), Department of Radiology, Lucas Center (S.J.H., M.E.M., J.R.), and Department of Neurosurgery, Division of Pediatric Neurosurgery (G.A.G.), Stanford University, Stanford, Calif; Department of Radiology, Pediatric MRI and CT, Division of Pediatric Radiology, Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Room G516, Palo Alto, CA 94304 (M.I., N.N.N., S.N., Y.Z., K.W.Y.); and Department of Neurosurgery, Division of Pediatric Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT (S.H.C.). From the 2018 RSNA Annual Meeting
| |
Collapse
|
10
|
Gao J, Chen X, Ma T, He B, Li P, Zhao Y, Ma Y, Zhuang J, Yin Y. PEG-Ceramide Nanomicelles Induce Autophagy and Degrade Tau Proteins in N2a Cells. Int J Nanomedicine 2020; 15:6779-6789. [PMID: 32982233 PMCID: PMC7494393 DOI: 10.2147/ijn.s258311] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose Alzheimer’s disease (AD) is a neurodegenerative disorder that manifests as abnormal behavior and a progressive decline in memory. Although the pathogenesis of AD is due to the excessive deposition of amyloid β protein (Aβ) outside the neurons in the brain, evidence suggests that tau proteins may be a better target for AD therapy. In neurodegenerative diseases, a decrease in autophagy results in the failure to eliminate abnormally deposited or misfolded proteins. Therefore, induction of autophagy may be an effective way to eliminate tau proteins in the treatment of AD. We investigated the effects of polyethylene glycol (PEG)-ceramide nanomicelles on autophagy and on tau proteins in N2a, a murine neuroblastoma metrocyte cell line. Methods Ceramide is a sphingolipid bioactive molecule that induces autophagy. PEG-ceramide is a polymer that is composed of the hydrophobic chain of ceramide and the hydrophilic chain of PEG-2000. In this study, we prepared PEG-ceramide nanomicelles that were 10–20 nm in size and had nearly neutral zeta potential. Results The results show that PEG-ceramide nanomicelles caused an increase in the LC3-II/LC3-I ratio, while p62 protein levels decreased. Confocal microscopy revealed a significant increase in the number of dots corresponding to autophagosomes and autolysosomes, which indicated autophagic activation. Moreover, PEG-ceramide nanomicelles induced tau degradation in N2a cells through autophagy. Conclusion In summary, we have confirmed that PEG-ceramide nanomicelles enhanced autophagic flux and degraded overexpressed human tau proteins in N2a cells by regulating the autophagy pathway. Thus, PEG-ceramide nanomicelles show great promise as agents to induce autophagy and degrade tau proteins in the treatment of AD.
Collapse
Affiliation(s)
- Jie Gao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China.,Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Xiaohan Chen
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Tianjun Ma
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Bin He
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Peng Li
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Yucheng Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Yuejin Ma
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - Jianhua Zhuang
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| | - You Yin
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People's Republic of China
| |
Collapse
|
11
|
Cancer stem cells and ceramide signaling: the cutting edges of immunotherapy. Mol Biol Rep 2020; 47:8101-8111. [PMID: 32885363 DOI: 10.1007/s11033-020-05790-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022]
Abstract
The multipotent, self renewing "cancer stem cells" (CSCs), a small population within tumor microenvironment facilitates transformed cells to grow and propagate within the body. The CSCs are discovered as resistant to the chemotherapeutic drug with distinct immunological characteristics. In recent years, immunologically targeting CSCs have emerged as an integral part of effective and successful cancer therapy. CSCs notably exhibit dysregulation in conventional sub-cellular sphingolipid metabolism. Recently, ceramide decaying enzymes have been shown to activate alternative ceramide signaling pathways leading to reduction in efficacy of the chemotherapeutic drugs. Therefore, a control over ceramide mediated modulations of CSCs offers an attractive dimension of effective cancer treatment strategy in future. In this review, we focused on the recent findings on broad spectrum of ceramide mediated signaling in CSCs within the tumor niche and their role in potential cancer immunotherapy.
Collapse
|
12
|
Ordóñez-Gutiérrez L, Wandosell F. Nanoliposomes as a Therapeutic Tool for Alzheimer's Disease. Front Synaptic Neurosci 2020; 12:20. [PMID: 32523525 PMCID: PMC7261886 DOI: 10.3389/fnsyn.2020.00020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
The accumulation of extracellular amyloid-beta (Aβ), denoted as senile plaques, and intracellular neurofibrillary tangles (formed by hyperphosphorylated Tau protein) in the brain are two major neuropathological hallmarks of Alzheimer's disease (AD). The current and most accepted hypothesis proposes that the oligomerization of Aβ peptides triggers the polymerization and accumulation of amyloid, which leads to the senile plaques. Several strategies have been reported to target Aβ oligomerization/polymerization. Since it is thought that Aβ levels in the brain and peripheral blood maintain equilibrium, it has been hypothesized that enhancing peripheral clearance (by shifting this equilibrium towards the blood) might reduce Aβ levels in the brain, known as the sink effect. This process has been reported to be effective, showing a reduction in Aβ burden in the brain as a consequence of the peripheral reduction of Aβ levels. Nanoparticles (NPs) may have difficulty crossing the blood-brain barrier (BBB), initially due to their size. It is not clear whether particles in the range of 50-100 nm should be able to cross the BBB without being specifically modified for it. Despite the size limitation of crossing the BBB, several NP derivatives may be proposed as therapeutic tools. The purpose of this review is to summarize some therapeutic approaches based on nanoliposomes using two complementary examples: First, unilamellar nanoliposomes containing Aβ generic ligands, such as sphingolipids, gangliosides or curcumin, or some sphingolipid bound to the binding domain of ApoE; and second, nanoliposomes containing monoclonal antibodies against Aβ. Following similar rationale NPs of poly(lactide-co-glycolide)-poly (ethylene glycol) conjugated with curcumin-derivate (PLGA-PEG-B6/Cur) were reported to improve the spatial learning and memory capability of APP/PS1 mice, compared with native curcumin treatment. Also, some new nanostructures such as exosomes have been proposed as a putative therapeutic and prevention strategies of AD. Although the unquestionable interest of this issue is beyond the scope of this review article. The potential mechanisms and significance of nanoliposome therapies for AD, which are still are in clinical trials, will be discussed.
Collapse
Affiliation(s)
- Lara Ordóñez-Gutiérrez
- Department of Molecular Neurobiology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Department of Molecular Neurobiology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
13
|
|
14
|
Suhrland C, Truman J, Obeid LM, Sitharaman B. Delivery of long chain C16and C24ceramide in HeLa cells using oxidized graphene nanoribbons. J Biomed Mater Res B Appl Biomater 2019; 108:1141-1156. [DOI: 10.1002/jbm.b.34465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/24/2019] [Accepted: 07/13/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Cassandra Suhrland
- Department of Biomedical EngineeringStony Brook University Stony Brook New York
| | - Jean‐Philip Truman
- Department of Medicine and the Stony Brook Cancer Center, Health Science CenterStony Brook University Stony Brook New York
| | - Lina M. Obeid
- Department of Medicine and the Stony Brook Cancer Center, Health Science CenterStony Brook University Stony Brook New York
| | - Balaji Sitharaman
- Department of Biomedical EngineeringStony Brook University Stony Brook New York
| |
Collapse
|
15
|
Lai M, La Rocca V, Amato R, Freer G, Pistello M. Sphingolipid/Ceramide Pathways and Autophagy in the Onset and Progression of Melanoma: Novel Therapeutic Targets and Opportunities. Int J Mol Sci 2019; 20:ijms20143436. [PMID: 31336922 PMCID: PMC6678284 DOI: 10.3390/ijms20143436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a malignant tumor deriving from neoplastic transformation of melanocytes. The incidence of melanoma has increased dramatically over the last 50 years. It accounts for most cases of skin cancer deaths. Early diagnosis leads to remission in 90% of cases of melanoma; conversely, for melanoma at more advanced stages, prognosis becomes more unfavorable also because dvanced melanoma is often resistant to pharmacological and radiological therapies due to genetic plasticity, presence of cancer stem cells that regenerate the tumor, and efficient elimination of drugs. This review illustrates the role of autophagy in tumor progression and resistance to therapy, focusing on molecular targets for future drugs.
Collapse
Affiliation(s)
- Michele Lai
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Rachele Amato
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Giulia Freer
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Mauro Pistello
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy.
- Virology Unit, Pisa University Hospital, 56127 Pisa, Italy.
| |
Collapse
|
16
|
Guo X, Dong C, Liu Q, Zhu X, Zuo S, Zhang H. The sustained and targeted treatment of hemangiomas by propranolol-loaded CD133 aptamers conjugated liposomes-in-microspheres. Biomed Pharmacother 2019; 114:108823. [PMID: 30965238 DOI: 10.1016/j.biopha.2019.108823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 01/07/2023] Open
Abstract
We previously developed propranolol-encapsulated liposomes-in-microspheres (PLIM) to realize the sustained propranolol release for the treatment of hemangiomas. However, the liposomes released from the microspheres still lacked specificity for CD133-positive hemangioma-derived stem cells (HemSCs) which are considered to be the seeds of hemangiomas. Therefore, we hereby encapsulated propranolol-loaded CD133 aptamers conjugated liposomes in poly(lactic-co-glycolic acid (PLGA) microspheres to develop propranolol-loaded CD133 aptamers conjugated liposomes-in-microspheres (PCLIM), to realize the aim of the sustained and targeted therapy of hemangiomas. The evaluation of the release of propranolol from PCLIM was carried out, and the cytotoxic effect and angiogenic growth factor expression inhibitory ability of PCLIM were performed in HemSCs. The in vivo hemangioma inhibitory ability of PCLIM was also investigated in nude mice with subcutaneous human hemangiomas. PCLIM possessed a desired size of 29.2 μm, drug encapsulation efficiency (25.3%), and a prolonged drug release for 40 days. Importantly, PCLIM could inhibit HemSCs proliferation and the protein expression of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor-A (VEGF) in HemSCs to a greater extent compared with PLIM. In nude mice bearing hemangioma xenograft, PCLIM showed the best therapeutic efficacy towards hemangiomas, as reflected by remarkably decreased hemangioma volume, weight and microvessel density (MVD). Thus, our results demonstrated that PCLIM realized the sustained and targeted treatment of hemangiomas, resulting in remarkable inhibition of hemangiomas.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Hemangioma & Vascular Malformation, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Changxian Dong
- Department of Hemangioma & Vascular Malformation, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China.
| | - Qiuyu Liu
- Department of Pathology, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Xiaoshuang Zhu
- Department of Hemangioma & Vascular Malformation, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Song Zuo
- Department of Hemangioma & Vascular Malformation, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Hongyu Zhang
- Department of Hemangioma & Vascular Malformation, The People's Hospital of Zhengzhou University, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| |
Collapse
|
17
|
Choudhury H, Pandey M, Yin TH, Kaur T, Jia GW, Tan SQL, Weijie H, Yang EKS, Keat CG, Bhattamishra SK, Kesharwani P, Md S, Molugulu N, Pichika MR, Gorain B. Rising horizon in circumventing multidrug resistance in chemotherapy with nanotechnology. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:596-613. [PMID: 31029353 DOI: 10.1016/j.msec.2019.04.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/24/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is one of the key barriers in chemotherapy, leading to the generation of insensitive cancer cells towards administered therapy. Genetic and epigenetic alterations of the cells are the consequences of MDR, resulted in drug resistivity, which reflects in impaired delivery of cytotoxic agents to the cancer site. Nanotechnology-based nanocarriers have shown immense shreds of evidence in overcoming these problems, where these promising tools handle desired dosage load of hydrophobic chemotherapeutics to facilitate designing of safe, controlled and effective delivery to specifically at tumor microenvironment. Therefore, encapsulating drugs within the nano-architecture have shown to enhance solubility, bioavailability, drug targeting, where co-administered P-gp inhibitors have additionally combat against developed MDR. Moreover, recent advancement in the stimuli-sensitive delivery of nanocarriers facilitates a tumor-targeted release of the chemotherapeutics to reduce the associated toxicities of chemotherapeutic agents in normal cells. The present article is focused on MDR development strategies in the cancer cell and different nanocarrier-based approaches in circumventing this hurdle to establish an effective therapy against deadliest cancer disease.
Collapse
Affiliation(s)
- Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia.
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Tan Hui Yin
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Taasjir Kaur
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Gan Wei Jia
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - S Q Lawrence Tan
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - How Weijie
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Eric Koh Sze Yang
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Chin Guan Keat
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Subrat Kumar Bhattamishra
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nagasekhara Molugulu
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mallikarjuna Rao Pichika
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia; Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor 47500, Malaysia.
| |
Collapse
|
18
|
Nanodelivery systems for overcoming limited transportation of therapeutic molecules through the blood-brain barrier. Future Med Chem 2018; 10:2659-2674. [PMID: 30499740 DOI: 10.4155/fmc-2018-0208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Due to the impermeable structure and barrier function of the blood-brain barrier (BBB), the delivery of therapeutic molecules into the CNS is extremely limited. Nanodelivery systems are regarded as the most effective and versatile carriers for the CNS, as they can transport cargo molecules across the BBB via various mechanisms. This review emphasizes the multi-functionalization strategies of nanodelivery systems and combinatorial approaches for the delivery of therapeutic drugs and genes into the CNS. The characteristics and functions of the BBB and underlying mechanisms of molecular translocation across the BBB are also described.
Collapse
|
19
|
Chen D, Pan X, Xie F, Lu Y, Zou H, Yin C, Zhang Y, Gao J. Codelivery of doxorubicin and elacridar to target both liver cancer cells and stem cells by polylactide-co-glycolide/d-alpha-tocopherol polyethylene glycol 1000 succinate nanoparticles. Int J Nanomedicine 2018; 13:6855-6870. [PMID: 30498347 PMCID: PMC6207383 DOI: 10.2147/ijn.s181928] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose Liver cancer is the third leading cause of cancer-related deaths worldwide. Liver cancer stem cells (LCSCs) are a subpopulation of cancer cells that are responsible for the initiation, progression, drug resistance, recurrence, and metastasis of liver cancer. Recent studies have suggested that the eradication of both LCSCs and liver cancer cells is necessary because the conversion of cancer stem cells (CSCs) to cancer cells occasionally occurs. As ATP-binding cassette (ABC) transporters are overexpressed in both CSCs and cancer cells, combined therapies using ABC transporter inhibitors and chemotherapy drugs could show superior therapeutic efficacy in liver cancer. In this study, we developed poly(lactide-co-glycolide)/d-alpha-tocopherol polyethylene glycol 1000 succinate nanoparticles to accomplish the simultaneous delivery of an optimized ratio of doxorubicin (DOX) and elacridar (ELC) to target both LCSCs and liver cancer cells. Methods Median-effect analysis was used for screening of DOX and ELC for synergy in liver cancer cells (HepG2 cells) and LCSCs (HepG2 tumor sphere [HepG2-TS]). Then, nanoparticles loaded with DOX and ELC at the optimized ratio (NDEs) were prepared by nanoprecipitation method. The cytotoxicity and colony and tumor sphere formation ability of nanoparticles were investigated in vitro, and the tissue distribution and antitumor activity of nanoparticles were evaluated in vivo. Results We demonstrated that a DOX/ELC molar ratio of 1:1 was synergistic in HepG2 cells and HepG2-TS. NDEs were shown to exhibit significantly increased cytotoxic effects against both HepG2 and HepG2-TS compared with DOX-loaded nanoparticles (NDs) or ELC-loaded nanoparticles (NEs) in vitro. In vivo studies demonstrated that the nanoparticles exhibited better tumor targeting, with NDE showing the strongest antitumor activity with lower systemic toxicity. Conclusion These results suggested that NDE represented a promising combination therapy against liver cancer by targeting both liver cancer cells and CSCs.
Collapse
Affiliation(s)
- Dazhong Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China, .,Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China, .,Department of Planning, Kunming General Hospital of Chengdu Military Command, Yunnan 650032, China
| | - Xiaoli Pan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| | - Hao Zou
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei 441000, China,
| | - Jie Gao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China, .,Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| |
Collapse
|
20
|
Wang T, Feng L, Yang S, Liu Y, Zhang N. Ceramide lipid-based nanosuspension for enhanced delivery of docetaxel with synergistic antitumor efficiency. Drug Deliv 2017; 24:800-810. [PMID: 28502199 PMCID: PMC8241063 DOI: 10.1080/10717544.2016.1225853] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ceramide (CE), a bioactive lipid with tumor suppression, has been widely used as a drug carrier and enhancer for cancer therapy. CE-based combination therapy was prone to be attractive in cancer therapy. In our previous study, the combination of CE and docetaxel (DTX) was proved to be an effective strategy for cancer therapy. To further improve the antitumor efficiency of DTX, the CE lipid-based nanosuspensions (LNS) was prepared for the delivery of DTX to exhibit synergistic therapeutic effect. The enhanced delivery and synergistic therapeutic effect of DTX-loaded CE-LNS (CE + DTX-LNS) were evaluated. CE + DTX-LNS exhibited spherical or ellipsoidal shape, uniform particle size distribution (108.1 ± 3.8 nm), sustained release characteristics and good stability in vitro. Notably, CE + DTX-LNS could effectively co-localize CE and DTX into same tumor cell and subsequently play synergistic cell damage effect compared with CE-LNS + DTX-LNS (p < 0.05). The in vivo fluorescence imaging results showed that CE + DTX-LNS could effectively prolong the in vivo circulation time and enhance the accumulation in tumor sites. Moreover, the antitumor efficacy of CE + DTX-LNS observed in B16 murine melanoma model was 93.94 ± 2.77%, significantly higher than that of CE-LNS, DTX-LNS, Duopafei® (p < 0.01) and CE-LNS + DTX-LNS (p < 0.05), respectively, demonstrating that co-delivery of CE and DTX into same tumor cell was the basis for enhanced synergistic therapeutic effect. Furthermore, histological examination of Blank-LNS showed no visible tissue toxicity compared to normal saline. Consequently, CE-LNS could effectively delivery DTX and CE + DTX-LNS exhibit synergistic inhibition of tumor growth due to the co-localization of CE and DTX. CE-LNS hold great potential to be an appropriate carrier for CE-based combination chemotherapy.
Collapse
Affiliation(s)
- Tianqi Wang
- a School of Pharmaceutical Science, Shandong University , Ji'nan , People's Republic of China
| | - Lixia Feng
- a School of Pharmaceutical Science, Shandong University , Ji'nan , People's Republic of China
| | - Shaomei Yang
- a School of Pharmaceutical Science, Shandong University , Ji'nan , People's Republic of China
| | - Yongjun Liu
- a School of Pharmaceutical Science, Shandong University , Ji'nan , People's Republic of China
| | - Na Zhang
- a School of Pharmaceutical Science, Shandong University , Ji'nan , People's Republic of China
| |
Collapse
|
21
|
De Simone U, Caloni F, Gribaldo L, Coccini T. Human Co-culture Model of Neurons and Astrocytes to Test Acute Cytotoxicity of Neurotoxic Compounds. Int J Toxicol 2017; 36:463-477. [PMID: 29153031 DOI: 10.1177/1091581817739428] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alternative methods and their use in planning and conducting toxicology experiments have become essential for modern toxicologists, thus reducing or replacing living animals. Although in vitro human co-culture models allow the establishment of biologically relevant cell-cell interactions that recapitulate the tissue microenvironment and better mimic its physiology, the number of publications is limited specifically addressing this scientific area and utilizing this test method which could provide an additional valuable model in toxicological studies. In the present study, an in vitro model based on central nervous system (CNS) cell co-cultures was implemented using a transwell system combining human neuronal cells (SH-SY5Y cell line) and glial cells, namely astrocytes (D384 cell line), to investigate neuroprotection of D384 on SH-SY5Y and vice versa. The model was applied to test acute (24-48 hours) cytotoxicity of 3 different neurotoxicants: (1) methyl mercury (1-2.5 μM), (2) Fe3O4 nanoparticles (1-100 μg/mL), and (3) methylglyoxal (0.5-1 mM). Data were compared to mono-cultures evaluating the mitochondrial function and cell morphology. The results clearly showed that all compounds tested affected the mitochondrial activity and cell morphology in both mono-culture and co-culture conditions. However, astrocytes, when cultured together with neurons, diminish the neurotoxicant-induced cytotoxic effects that occurred in neurons cultured alone, and astrocytes become more resistant in the presence of neurons. This human CNS co-culture system seems a suitable cell model to feed high-throughput acute screening platforms and to evaluate both human neuronal and astrocytic toxicity and neuroprotective effects of new and emerging materials (eg, nanomaterials) and new products with improved sensitivity due to the functional neuron-astrocyte metabolic interactions.
Collapse
Affiliation(s)
- Uliana De Simone
- 1 Laboratory of Experimental and Clinical Toxicology, Toxicology Unit, ICS Maugeri SpA-Benefit Corporation, IRCCS Pavia, Pavia, Italy
| | - Francesca Caloni
- 2 Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Milano, Italy
| | - Laura Gribaldo
- 3 European Commission, Directorate General Joint Research Centre, Directorate F-Health, Consumers and Reference Materials, Chemicals Safety and Alternative Methods Unit, Ispra, Italy
| | - Teresa Coccini
- 1 Laboratory of Experimental and Clinical Toxicology, Toxicology Unit, ICS Maugeri SpA-Benefit Corporation, IRCCS Pavia, Pavia, Italy
| |
Collapse
|
22
|
Guo X, Zhu X, Liu D, Gong Y, Sun J, Dong C. Continuous delivery of propranolol from liposomes-in-microspheres significantly inhibits infantile hemangioma growth. Int J Nanomedicine 2017; 12:6923-6936. [PMID: 29075111 PMCID: PMC5609781 DOI: 10.2147/ijn.s137634] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose To reduce the adverse effects and high frequency of administration of propranolol to treat infantile hemangioma, we first utilized propranolol-loaded liposomes-in-microsphere (PLIM) as a novel topical release system to realize sustained release of propranolol. Methods PLIM was developed from encapsulating propranolol-loaded liposomes (PLs) in microspheres made of poly(lactic-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) copolymers (PLGA-PEG-PLGA). The release profile of propranolol from PLIM was evaluated, and its biological activity was investigated in vitro using proliferation assays on hemangioma stem cells (HemSCs). Tumor inhibition was studied in nude mice bearing human subcutaneous infantile hemangioma. Results The microspheres were of desired particle size (~77.8 μm) and drug encapsulation efficiency (~23.9%) and achieved sustained drug release for 40 days. PLIM exerted efficient inhibition of the proliferation of HemSCs and significantly reduced the expression of two angiogenesis factors (vascular endothelial growth factor-A [VEGF-A] and basic fibroblast growth factor [bFGF]) in HemSCs. Notably, the therapeutic effect of PLIM in hemangioma was superior to that of propranolol and PL in vivo, as reflected by significantly reduced hemangioma volume, weight, and microvessel density. The mean hemangioma weight of the PLIM-treated group was significantly lower than that of other groups (saline =0.28 g, propranolol =0.21 g, PL =0.13 g, PLIM =0.03 g; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.001, PLIM vs PL: P<0.001). The mean microvessel density of the PLIM-treated group was significantly lower than that of other groups (saline =40 vessels/mm2, propranolol =31 vessels/mm2, PL =25 vessels/mm2, PLIM =11 vessels/mm2; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.01, PLIM vs PL: P<0.05). Conclusion Our findings show that PLIM is a very promising approach to locally and efficiently deliver propranolol to the hemangioma site leading to a significant inhibition of infantile hemangioma.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Xiaoshuang Zhu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Dakan Liu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Yubin Gong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Jing Sun
- Department of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Changxian Dong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
23
|
Øverbye A, Holsæter AM, Markus F, Škalko-Basnet N, Iversen TG, Torgersen ML, Sønstevold T, Engebraaten O, Flatmark K, Mælandsmo GM, Skotland T, Sandvig K. Ceramide-containing liposomes with doxorubicin: time and cell-dependent effect of C6 and C12 ceramide. Oncotarget 2017; 8:76921-76934. [PMID: 29100358 PMCID: PMC5652752 DOI: 10.18632/oncotarget.20217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/17/2017] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin, a widely used chemotherapeutic drug, has several potential high-risk side effects including cardiomyopathy. Furthermore, cellular resistance to this drug develops with time. By using liposomes as carrier vesicles both the side effects and drug resistance might be avoided. In this study we have investigated the cytotoxic effect of doxorubicin encapsulated in liposomes with and without ceramides containing 6 or 12 carbon atoms in the N-amidated fatty acyl chains. The short-chain ceramide species were included in the liposomal compositions due to their pro-apoptotic properties, which might cause a synergistic anticancer effect. We demonstrate that the ceramide species enhance the liposomal doxorubicin toxicity in a cell-specific manner. The C6-ceramide effect is most pronounced in cervical cancer cells (HeLa) and colon cancer cells (HCT116), whereas the C12-ceramide effect is strongest in breast cancer cells (MDA-MB-231). Moreover, the study reveals the importance of investigating cell toxicity at several time points and in different cell-lines, to assess drug-and formulation-induced cytotoxic effects in vitro. Furthermore, our data show that the cytotoxicity obtained with the nanocarriers in vitro, does not necessarily reflect their ability to inhibit tumor growth in vivo. We speculate that the larger effect of Caelyx® than our liposomes in vivo is due to a greater in vivo stability of Caelyx®.
Collapse
Affiliation(s)
- Anders Øverbye
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ann Mari Holsæter
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Fusser Markus
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Tore-Geir Iversen
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Tonje Sønstevold
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Olav Engebraaten
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gunhild Mari Mælandsmo
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway.,Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Tore Skotland
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol Res 2017; 126:2-30. [PMID: 28760489 DOI: 10.1016/j.phrs.2017.07.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/29/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
Multidrug resistance (MDR) is associated with a wide range of pathological changes at different cellular and intracellular levels. Nanoparticles (NPs) have been extensively exploited as the carriers of MDR reversing payloads to resistant tumor cells. However, when properly formulated in terms of chemical composition and physicochemical properties, NPs can serve as beyond delivery systems and help overcome MDR even without carrying a load of chemosensitizers or MDR reversing molecular cargos. Whether serving as drug carriers or beyond, a wise design of the nanoparticulate systems to overcome the cellular and intracellular alterations underlying the resistance is imperative. Within the current review, we will initially discuss the cellular changes occurring in resistant cells and how such changes lead to chemotherapy failure and cancer cell survival. We will then focus on different mechanisms through which nanosystems with appropriate chemical composition and physicochemical properties can serve as MDR reversing units at different cellular and intracellular levels according to the changes that underlie the resistance. Finally, we will conclude by discussing logical grounds for a wise and rational design of MDR reversing nanoparticulate systems to improve the cancer therapeutic approaches.
Collapse
Affiliation(s)
- Manu S Singh
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany
| | - Salma N Tammam
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Department of Pharmaceutical Technology, German University of Cairo, Egypt
| | | | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France.
| |
Collapse
|
25
|
Weil BD, Jenkins MJ, Uddin S, Bracewell DG, Wellings D, Farid SS, Veraitch F. An integrated experimental and economic evaluation of cell therapy affinity purification technologies. Regen Med 2017. [DOI: 10.2217/rme-2016-0156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim: To present an integrated techno-economic analysis assessing the feasibility of affinity purification technologies using the manufacture of induced pluripotent stem cell-derived progenitor photoreceptors for retinal dystrophies as a case study. Materials & methods: Sort purity, progenitor yield and viable cell recovery were investigated for three cell sorting techniques: fluorescent-activated cell sorting (FACS); magnetic-activated cell sorting (MACS); and a novel technology SpheriTech beads. Experimentally derived metrics were incorporated into an advanced bioprocess economics tool to determine cost of goods per dose for each technology. Results & conclusion: Technical and bioprocess benefits were noted with SpheriTech beads which, unlike FACS and MACS, require no cell labeling. This simplifies the bioprocess, reduces cell loss and leaves target cells label free. The economic tool predicted cost drivers and a critical dose (7 × 107 cells per dose) shifting the most cost-effective technology from FACS to MACS. Process optimization is required for SpheriTech to compete economically.
Collapse
Affiliation(s)
- Benjamin D Weil
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
- Royal Free Hospital Campus, Department of Haematology, University College London, Fleet Road, London NW3 2QG, UK
| | - Michael J Jenkins
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Siddique Uddin
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Daniel G Bracewell
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Donald Wellings
- SpheriTech Ltd, The Heath Business & Technical Park, Runcorn, Cheshire WA7 4QX, UK
| | - Suzanne S Farid
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| | - Farlan Veraitch
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
26
|
Wang M, Xie F, Wen X, Chen H, Zhang H, Liu J, Zhang H, Zou H, Yu Y, Chen Y, Sun Z, Wang X, Zhang G, Yin C, Sun D, Gao J, Jiang B, Zhong Y, Lu Y. Therapeutic PEG-ceramide nanomicelles synergize with salinomycin to target both liver cancer cells and cancer stem cells. Nanomedicine (Lond) 2017; 12:1025-1042. [PMID: 28440698 DOI: 10.2217/nnm-2016-0408] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Salinomycin (SAL)-loaded PEG-ceramide nanomicelles (SCM) were prepared to target both liver cancer cells and cancer stem cells. MATERIALS & METHODS The synergistic ratio of SAL/PEG-ceramide was evaluated to prepare SCM, and the antitumor activity of SCM was examined both in vitro and in vivo. RESULTS SAL/PEG-ceramide molar ratio of 1:4 was chosen as the synergistic ratio, and SCM showed superior cytotoxic effect and increased apoptosis-inducing activity in both liver cancer cells and cancer stem cells. In vivo, SCM showed the best tumor inhibitory effect with a safety profile. CONCLUSION Thus, PEG-ceramide nanomicelles could serve as an effective and safe therapeutic drug carrier to deliver SAL into liver cancer, opening up the avenue of using PEG-ceramide as therapeutic drug carriers.
Collapse
Affiliation(s)
- Meiping Wang
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Fangyuan Xie
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Xikai Wen
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Han Chen
- Department of General Surgery, 411 Hospital of Chinese People's Liberation Army, 15 East Jiangwan Road, Shanghai 200081, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Junjie Liu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - He Zhang
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Hao Zou
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yuan Yu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yan Chen
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhiguo Sun
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xinxia Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Guoqing Zhang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Gao
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Beige Jiang
- Third Department of HepaticSurgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Yanqiang Zhong
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
27
|
Gao J, Ochyl LJ, Yang E, Moon JJ. Cationic liposomes promote antigen cross-presentation in dendritic cells by alkalizing the lysosomal pH and limiting the degradation of antigens. Int J Nanomedicine 2017; 12:1251-1264. [PMID: 28243087 PMCID: PMC5317250 DOI: 10.2147/ijn.s125866] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cationic liposomes (CLs) have been widely examined as vaccine delivery nanoparticles since they can form complexes with biomacromolecules, promote delivery of antigens and adjuvant molecules to antigen-presenting cells (APCs), and mediate cellular uptake of vaccine components. CLs are also known to trigger antigen cross-presentation - the process by which APCs internalize extracellular protein antigens, degrade them into minimal CD8+ T-cell epitopes, and present them in the context of major histocompatibility complex-I (MHC-I). However, the precise mechanisms behind CL-mediated induction of cross-presentation and cross-priming of CD8+ T-cells remain to be elucidated. In this study, we have developed two distinct CL systems and examined their impact on the lysosomal pH in dendritic cells (DCs), antigen degradation, and presentation of peptide:MHC-I complexes to antigen-specific CD8+ T-cells. To achieve this, we have used 3β-[N-(N',N'-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol) and 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) as the prototypical components of CLs with tertiary amine groups and compared the effect of CLs and anionic liposomes on lysosomal pH, antigen degradation, and cross-presentation by DCs. Our results showed that CLs, but not anionic liposomes, elevated the lysosomal pH in DCs and reduced antigen degradation, thereby promoting cross-presentation and cross-priming of CD8+ T-cell responses. These studies shed new light on CL-mediated cross-presentation and suggest that intracellular fate of vaccine components and subsequent immunological responses can be controlled by rational design of nanomaterials.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA; Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China; Biointerfaces Institute
| | - Lukasz J Ochyl
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute
| | | | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Tian B, Liu R, Chen S, Chen L, Liu F, Jia G, Dong Y, Li J, Chen H, Lu J. Mannose-coated gadolinium liposomes for improved magnetic resonance imaging in acute pancreatitis. Int J Nanomedicine 2017; 12:1127-1141. [PMID: 28260882 PMCID: PMC5325132 DOI: 10.2147/ijn.s123290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Acute pancreatitis (AP) is an acute inflammatory condition of the pancreas. The symptoms, treatment, and prognosis of mild and severe AP are different, and severe AP is a potentially life-threatening disease with a high incidence of complications and high mortality rate. Thus, it is urgent to develop an effective approach to reliably discriminate between mild and severe AP. Methods We have developed novel gadolinium-diethylenetriaminepentaacetic (Gd-DTPA)-loaded mannosylated liposomes (named thereafter M-Gd-NL) that preferably target macrophages in AP. The targeting ability of M-Gd-NL toward macrophages in AP and its ability to discriminate between mild and severe AP were evaluated. Results The liposomes were of desired particle size (~100 nm), Gd-DTPA encapsulation efficiency (~85%), and stability. M-Gd-NL and non-targeted Gd-DTPA-loaded liposomes (Gd-NL) exhibited increased relaxivity compared with Gd-DTPA. Compared with Gd-NL and Gd-DTPA, M-Gd-NL showed increased uptake in macrophages, resulting in increased T1 imaging ability both in vitro (macrophage cell line) and in vivo (severe AP model). Importantly, M-Gd-NL had the ability to discriminate between mild and severe AP, as reflected by a significantly higher T1 magnetic resonance imaging signal in severe AP than in mild AP. M-Gd-NL did not show severe organ toxicity in rats. Conclusion Our data suggest that M-Gd-NL had enhanced magnetic resonance imaging ability by targeting macrophages in AP and good ability to discriminate between mild and severe AP. We believe that M-Gd-NL could shed new light on the diagnosis of AP in the near future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Huaiwen Chen
- Center of Clinical and Translational Medicine, Shanghai Changhai Hospital, The Second Military Medical University; Sunlipo Biotech Research Center for Nanomedicine, Shanghai, People's Republic of China
| | | |
Collapse
|
29
|
Chen D, Xie F, Sun D, Yin C, Gao J, Zhong Y. Nanomedicine-Mediated Combination Drug Therapy in Tumor. ACTA ACUST UNITED AC 2017. [DOI: 10.2174/1874844901704010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background:Combined chemotherapy has gradually become one of the conventional methods of cancer treatment due to the limitation of monotherapy. However, combined chemotherapy has several drawbacks that may lead to treatment failure because drug synergy cannot be guaranteed, achievement of the optimal synergistic drug ratio is difficult, and drug uptake into the tumor is inconsistent. Nanomedicine can be a safe and effective form of drug delivery, which may address the problems associated with combination chemotherapy.Objective:This review summarizes the recent research in this area, including the use of nanoparticles, liposomes, lipid-polymer hybrid nanoparticles, and polymeric micelles, and provides new approach for combined chemotherapy.Methods:By collecting and referring to the related literature in recent years.Results:Compared with conventional drugs, nanomedicine has the following advantages: it increases bioavailability of poorly soluble drugs, prolongs drug circulation timein vivo, and permits multiple drug loading, all of which could improve drug efficacy and reduce toxicity. Furthermore, nanomedicine can maintain the synergistic ratio of the drugs; deliver the drugs to the tumor at the same time, such that two or more drugs of tumor treatment achieve synchronization in time and space; and alter the pharmacokinetics and distribution profilein vivosuch that these are dependent on nanocarrier properties (rather than being dependent on the drugs themselves).Conclusion:Therefore, nanomedicine-mediated combination drug therapy is promising in the treatment of tumors.
Collapse
|
30
|
Zhu M, Chen S, Hua L, Zhang C, Chen M, Chen D, Dong Y, Zhang Y, Li M, Song X, Chen H, Zheng H. Self-targeted salinomycin-loaded DSPE-PEG-methotrexate nanomicelles for targeting both head and neck squamous cell carcinoma cancer cells and cancer stem cells. Nanomedicine (Lond) 2017; 12:295-315. [PMID: 28093940 DOI: 10.2217/nnm-2016-0382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To target both head and neck squamous cell carcinoma (HNSCC) cells and cancer stem cells (CSCs) by salinomycin-loaded DSPE-PEG-MTX (synthesized using DSPE-PEG2000-NH2 and methotrexate) nanomicelles (M-SAL-MTX). MATERIALS & METHODS The characterization, antitumor activity and mechanism of M-SAL-MTX were evaluated. RESULTS & CONCLUSION M-SAL-MTX showed enhanced inhibitory effect toward both HNSCC CSCs and non-CSCs compared with a single treatment of methotrexate and salinomycin. In nude mice-bearing HNSCC xenografts, M-SAL-MTX suppressed tumor growth more effectively than other controls including combination of methotrexate and salinomycin. Therefore, M-SAL-MTX may provide a strategy for treating HNSCC by targeting both HNSCC CSCs and HNSCC cells.
Collapse
Affiliation(s)
- Minhui Zhu
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Shicai Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Libo Hua
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Caiyun Zhang
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Mengjie Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Donghui Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yinmei Dong
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yingying Zhang
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Meng Li
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xianmin Song
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huaiwen Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China.,Sunlipo Biotech Research Center for Nanomedicine, 3688 Tingwei Road, Shanghai 201507, China
| | - Hongliang Zheng
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
31
|
Gao J, Li W, Guo Y, Feng SS. Nanomedicine strategies for sustained, controlled and targeted treatment of cancer stem cells. Nanomedicine (Lond) 2016; 11:3261-3282. [PMID: 27854161 DOI: 10.2217/nnm-2016-0261] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cancer stem cells (CSCs) are original cancer cells that are of characteristics associated with normal stem cells. CSCs are toughest against various treatments and thus responsible for cancer metastasis and recurrence. Therefore, development of specific and effective treatment of CSCs plays a key role in improving survival and life quality of cancer patients, especially those in the metastatic stage. Nanomedicine strategies, which include prodrugs, micelles, liposomes and nanoparticles of biodegradable polymers, could substantially improve the therapeutic index of conventional therapeutics due to its manner of sustained, controlled and targeted delivery of high transportation efficiency across the cell membrane and low elimination by intracellular autophagy, and thus provide a practical solution to solve the problem encountered in CSCs treatment. This review gives briefly the latest information to summarize the concept, strategies, mechanisms and current status as well as future promises of nanomedicine strategies for treatment of CSCs.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, the Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wei Li
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Yajun Guo
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Si-Shen Feng
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China.,Department of Chemical & Biomolecular Engineering, National University of Singapore, Block E5, 02-11, 4 Engineering Drive 4, Singapore 117576, Singapore.,Suzhou NanoStar Biopharm Inc. Ltd, BioBay, Bld B2, Unit 604, 218 Xing-Hu Street, Suzhou Industrial Park, Suzhou 215123, China
| |
Collapse
|
32
|
Yang K, Lu Y, Xie F, Zou H, Fan X, Li B, Li W, Zhang W, Mei L, Feng SS, Yin Y, Liu Y, Zhang H, Yin C, Zhong Y, Gao J. Cationic liposomes induce cell necrosis through lysosomal dysfunction and late-stage autophagic flux inhibition. Nanomedicine (Lond) 2016; 11:3117-3137. [PMID: 27819530 DOI: 10.2217/nnm-2016-0289] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM The application of cationic liposomes (CLs) as nonviral vectors is hampered by their cellular toxicity. Thus we aim to investigate the mechanisms underlying the cellular toxicity of CLs. MATERIALS & METHODS The effect of CLs on the autophagic flux, autophagosome-lysosome fusion, lysosome membrane permeabilization and cell necrosis of liver cells was investigated. RESULTS & CONCLUSION Our results reveal a novel mechanism of CL-induced cell necrosis involving the induction of lysosome membrane permeabilization and late-stage autophagic flux inhibition that resulted in cytoplasmic release of cathepsin B, mitochondrial dysfunction and reactive oxygen species production, which are the key mediators of cell necrosis. Our study is important for revealing the cellular toxicity of CLs and designing safer gene delivery systems.
Collapse
Affiliation(s)
- Kaixuan Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China.,Department of Oncology, Affiliated Hospital of Guizhou Medcial University & Guizhou Cancer Hospital, Guiyang 550025, Guizhou, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Fangyuan Xie
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Hao Zou
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xiaoyu Fan
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Bohua Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Wei Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Wei Zhang
- Department of Respiratory & Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Lin Mei
- Division of Life & Health Sciences, Tsinghua University Shenzhen Graduate School, Shenzhen 518055, China
| | - Si-Shen Feng
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China.,Department of Chemical & Biomolecular Engineering, National University of Singapore, Block E5, 02-11, 4 Engineering Drive 4, Singapore 117576, Singapore
| | - You Yin
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yan Liu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yanqiang Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jie Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
33
|
Petters C, Thiel K, Dringen R. Lysosomal iron liberation is responsible for the vulnerability of brain microglial cells to iron oxide nanoparticles: comparison with neurons and astrocytes. Nanotoxicology 2015; 10:332-42. [DOI: 10.3109/17435390.2015.1071445] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Charlotte Petters
- Center for Biomedical Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany,
- Center for Environmental Research and Sustainable Technology, Bremen, Germany, and
| | - Karsten Thiel
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials, Bremen, Germany
| | - Ralf Dringen
- Center for Biomedical Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany,
- Center for Environmental Research and Sustainable Technology, Bremen, Germany, and
| |
Collapse
|