1
|
Jiao W, Wen N, Wang S, Zhou G, Lu Q, Su Z, Wang X, Hu S, Xie Y, Zhang N, Liu X. Effect of surface modification on the distribution of magnetic nanorings in hepatocellular carcinoma and immune cells. J Mater Chem B 2024; 12:2628-2638. [PMID: 38376513 DOI: 10.1039/d3tb02560h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Magnetic nanomaterial-mediated magnetic hyperthermia is a localized heating treatment modality that has been applied to treat aggressive cancer in clinics. In addition to being taken up by tumor cells to function in cancer therapy, magnetic nanomaterials can also be internalized by immune cells in the tumor microenvironment, which may contribute to regulating the anti-tumor immune effects. However, there exists little studies on the distribution of magnetic nanomaterials in different types of cells within tumor tissue. Herein, ferrimagnetic vortex-domain iron oxide nanorings (FVIOs) with or without the liver-cancer-targeting peptide SP94 have been successfully synthesized as a model system to investigate the effect of surface modification of FVIOs (with or without SP94) on the distribution of tumor cells and different immune cells in hepatocellular carcinoma (HCC) microenvironment of a mouse. The distribution ratio of FVIO-SP94s in tumor cells was 1.3 times more than that of FVIOs. Immune cells in the liver tumor microenvironment took up fewer FVIO-SP94s than FVIOs. In addition, myeloid cells were found to be much more amenable than lymphoid cells in terms of their ability to phagocytose nanoparticles. Specifically, the distributions of FVIOs/FVIO-SP94s in tumor-associated macrophages, dendritic cells, and myeloid-derived suppressor cells were 13.8%/12%, 3.7%/0.9%, and 6.3%/1.2%, respectively. While the distributions of FVIOs/FVIO-SP94s in T cells, B cells, and natural killer cells were 5.5%/0.7%, 3.0%/0.7%, and 0.4%/0.3%, respectively. The results described in this article enhance our understanding of the distribution of nanomaterials in the tumor microenvironment and provide a strategy for rational design of magnetic hyperthermia agents that can effectively regulate anti-tumor immune effects.
Collapse
Affiliation(s)
- Wangbo Jiao
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Nana Wen
- School of Science and Chemical Engineering, Ningxia Institute of Science and Technology, Shizuishan, Ningxia 753000, China
| | - Siyao Wang
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Guxiang Zhou
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Qiaoyi Lu
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Zijun Su
- School of materials, Sun Yat-Sen University, Shen Zhen, Guangdong 529406, China
| | - Xinxin Wang
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Shuwei Hu
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Youbang Xie
- Department of Hematology and Rheumatology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, Qinghai 810007, China
| | - Nan Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaoli Liu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| |
Collapse
|
2
|
Baabu PRS, Kumar HK, Gumpu MB, Babu K J, Kulandaisamy AJ, Rayappan JBB. Iron Oxide Nanoparticles: A Review on the Province of Its Compounds, Properties and Biological Applications. MATERIALS (BASEL, SWITZERLAND) 2022; 16:ma16010059. [PMID: 36614400 PMCID: PMC9820855 DOI: 10.3390/ma16010059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 05/14/2023]
Abstract
Materials science and technology, with the advent of nanotechnology, has brought about innumerable nanomaterials and multi-functional materials, with intriguing yet profound properties, into the scientific realm. Even a minor functionalization of a nanomaterial brings about vast changes in its properties that could be potentially utilized in various applications, particularly for biological applications, as one of the primary needs at present is for point-of-care devices that can provide swifter, accurate, reliable, and reproducible results for the detection of various physiological conditions, or as elements that could increase the resolution of current bio-imaging procedures. In this regard, iron oxide nanoparticles, a major class of metal oxide nanoparticles, have been sweepingly synthesized, characterized, and studied for their essential properties; there are 14 polymorphs that have been reported so far in the literature. With such a background, this review's primary focus is the discussion of the different synthesis methods along with their structural, optical, magnetic, rheological and phase transformation properties. Subsequently, the review has been extrapolated to summarize the effective use of these nanoparticles as contrast agents in bio-imaging, therapeutic agents making use of its immune-toxicity and subsequent usage in hyperthermia for the treatment of cancer, electron transfer agents in copious electrochemical based enzymatic or non-enzymatic biosensors and bactericidal coatings over biomaterials to reduce the biofilm formation significantly.
Collapse
Affiliation(s)
- Priyannth Ramasami Sundhar Baabu
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Hariprasad Krishna Kumar
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
- Acrophase, Indian Institute of Technology Madras, Chennai 600 036, Tamil Nadu, India
| | - Manju Bhargavi Gumpu
- Department of Physics, National Institute of Technology, Tiruchirappalli 620 015, Tamil Nadu, India
| | - Jayanth Babu K
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
- School of Electrical & Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | | | - John Bosco Balaguru Rayappan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
- School of Electrical & Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
- Correspondence:
| |
Collapse
|
3
|
Methotrexate-Transferrin-Functionalized Fe(Salen)-Polypyrrole Nanocomposites for Targeted Photo-/Magneto-Thermal Cancer Treatments. JOURNAL OF COMPOSITES SCIENCE 2022. [DOI: 10.3390/jcs6050136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Designing multi-modal topical drug delivery nanocarriers using nano-hybrid particles has received significant interest in targeted cancer therapy. In this study, magnetic Fe(salen)-conducting copolymer nanocomposites based on our previous iron salt-free synthesis method are surface-functionalized with methotrexate and transferrin proteins. The nano-hybrids show near-infrared-/magnetic field-responsive hyperthermal activity in vitro, which can be extraordinarily useful in magnetically guidable local cancer targeting as a versatile multi-modal therapeutic drug delivery system.
Collapse
|
4
|
Kraus S, Khandadash R, Hof R, Nyska A, Sigalov E, Eltanani M, Rukenstein P, Rabinovitz R, Kassem R, Antebi A, Shalev O, Cohen-Erner M, Goss G, Cyjon A. Novel Nanoparticle-Based Cancer Treatment, Effectively Inhibits Lung Metastases and Improves Survival in a Murine Breast Cancer Model. Front Oncol 2021; 11:761045. [PMID: 34804962 PMCID: PMC8602876 DOI: 10.3389/fonc.2021.761045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Sarah Nanoparticles (SaNPs) are unique multicore iron oxide-based nanoparticles, developed for the treatment of advanced cancer, following standard care, through the selective delivery of thermal energy to malignant cells upon exposure to an alternating magnetic field. For their therapeutic effect, SaNPs need to accumulate in the tumor. Since the potential accumulation and associated toxicity in normal tissues are an important risk consideration, biodistribution and toxicity were assessed in naïve BALB/c mice. Therapeutic efficacy and the effect on survival were investigated in the 4T1 murine model of metastatic breast cancer. Toxicity evaluation at various timepoints did not reveal any abnormal clinical signs, evidence of alterations in organ function, nor histopathologic adverse target organ toxicity, even after a follow up period of 25 weeks, confirming the safety of SaNP use. The biodistribution evaluation, following SaNP administration, indicated that SaNPs accumulate mainly in the liver and spleen. A comprehensive pharmacokinetics evaluation, demonstrated that the total percentage of SaNPs that accumulated in the blood and vital organs was ~78%, 46%, and 36% after 4, 13, and 25 weeks, respectively, suggesting a time-dependent clearance from the body. Efficacy studies in mice bearing 4T1 metastatic tumors revealed a 49.6% and 70% reduction in the number of lung metastases and their relative size, respectively, in treated vs. control mice, accompanied by a decrease in tumor cell viability in response to treatment. Moreover, SaNP treatment followed by alternating magnetic field exposure significantly improved the survival rate of treated mice compared to the controls. The median survival time was 29 ± 3.8 days in the treated group vs. 21.6 ± 4.9 days in the control, p-value 0.029. These assessments open new avenues for generating SaNPs and alternating magnetic field application as a potential novel therapeutic modality for metastatic cancer patients.
Collapse
Affiliation(s)
| | | | | | - Abraham Nyska
- Toxicologic Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | - Glenwood Goss
- Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada
| | - Arnoldo Cyjon
- Department of Oncology, Shamir Medical Center, Zerifin, Israel
| |
Collapse
|
5
|
Damasco JA, Ravi S, Perez JD, Hagaman DE, Melancon MP. Understanding Nanoparticle Toxicity to Direct a Safe-by-Design Approach in Cancer Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2186. [PMID: 33147800 PMCID: PMC7692849 DOI: 10.3390/nano10112186] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Nanomedicine is a rapidly growing field that uses nanomaterials for the diagnosis, treatment and prevention of various diseases, including cancer. Various biocompatible nanoplatforms with diversified capabilities for tumor targeting, imaging, and therapy have materialized to yield individualized therapy. However, due to their unique properties brought about by their small size, safety concerns have emerged as their physicochemical properties can lead to altered pharmacokinetics, with the potential to cross biological barriers. In addition, the intrinsic toxicity of some of the inorganic materials (i.e., heavy metals) and their ability to accumulate and persist in the human body has been a challenge to their translation. Successful clinical translation of these nanoparticles is heavily dependent on their stability, circulation time, access and bioavailability to disease sites, and their safety profile. This review covers preclinical and clinical inorganic-nanoparticle based nanomaterial utilized for cancer imaging and therapeutics. A special emphasis is put on the rational design to develop non-toxic/safe inorganic nanoparticle constructs to increase their viability as translatable nanomedicine for cancer therapies.
Collapse
Affiliation(s)
- Jossana A. Damasco
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.A.D.); (J.D.P.); (D.E.H.)
| | - Saisree Ravi
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Joy D. Perez
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.A.D.); (J.D.P.); (D.E.H.)
| | - Daniel E. Hagaman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.A.D.); (J.D.P.); (D.E.H.)
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.A.D.); (J.D.P.); (D.E.H.)
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
6
|
Belderbos S, González-Gómez MA, Cleeren F, Wouters J, Piñeiro Y, Deroose CM, Coosemans A, Gsell W, Bormans G, Rivas J, Himmelreich U. Simultaneous in vivo PET/MRI using fluorine-18 labeled Fe 3O 4@Al(OH) 3 nanoparticles: comparison of nanoparticle and nanoparticle-labeled stem cell distribution. EJNMMI Res 2020; 10:73. [PMID: 32607918 PMCID: PMC7326875 DOI: 10.1186/s13550-020-00655-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown potential for treatment of different diseases. However, their working mechanism is still unknown. To elucidate this, the non-invasive and longitudinal tracking of MSCs would be beneficial. Both iron oxide-based nanoparticles (Fe3O4 NPs) for magnetic resonance imaging (MRI) and radiotracers for positron emission tomography (PET) have shown potential as in vivo cell imaging agents. However, they are limited by their negative contrast and lack of spatial information as well as short half-life, respectively. In this proof-of-principle study, we evaluated the potential of Fe3O4@Al(OH)3 NPs as dual PET/MRI contrast agents, as they allow stable binding of [18F]F- ions to the NPs and thus, NP visualization and quantification with both imaging modalities. RESULTS 18F-labeled Fe3O4@Al(OH)3 NPs (radiolabeled NPs) or mouse MSCs (mMSCs) labeled with these radiolabeled NPs were intravenously injected in healthy C57Bl/6 mice, and their biodistribution was studied using simultaneous PET/MRI acquisition. While liver uptake of radiolabeled NPs was seen with both PET and MRI, mMSCs uptake in the lungs could only be observed with PET. Even some initial loss of fluoride label did not impair NPs/mMSCs visualization. Furthermore, no negative effects on blood cell populations were seen after injection of either the NPs or mMSCs, indicating good biocompatibility. CONCLUSION We present the application of novel 18F-labeled Fe3O4@Al(OH)3 NPs as safe cell tracking agents for simultaneous PET/MRI. Combining both modalities allows fast and easy NP and mMSC localization and quantification using PET at early time points, while MRI provides high-resolution, anatomic background information and long-term NP follow-up, hereby overcoming limitations of the individual imaging modalities.
Collapse
Affiliation(s)
- Sarah Belderbos
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium
| | - Manuel Antonio González-Gómez
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Jens Wouters
- Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000, Leuven, Belgium
| | - Yolanda Piñeiro
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven/UZ Leuven, 3000, Leuven, Belgium
| | - An Coosemans
- Laboratory for Tumor Immunology and Immunotherapy, ImmunOvar Research Group, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000, Leuven, Belgium.,Department of Gynaecology and Obstetrics, UZ Leuven, 3000, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Jose Rivas
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
Kuchur OA, Tsymbal SA, Shestovskaya MV, Serov NS, Dukhinova MS, Shtil AA. Metal-derived nanoparticles in tumor theranostics: Potential and limitations. J Inorg Biochem 2020; 209:111117. [PMID: 32473483 DOI: 10.1016/j.jinorgbio.2020.111117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/19/2022]
Abstract
Initially, metal derived nanoparticles have been used exclusively as contrasting agents in magnetic resonance imaging. Today, green routes of chemical synthesis together with numerous modifications of the core and surface gave rise to a plethora of biomedical applications of metal derived nanoparticles including tumor imaging, diagnostics, and therapy. These materials are an emerging class of tools for tumor theranostics. Nevertheless, the spectrum of clinically approved metal nanoparticles remains narrow, as the safety, specificity and efficiency still have to be improved. In this review we summarize the major directions for development and biomedical applications of metal based nanoparticles and analyze their effects on tumor cells and microenvironment. We discuss the advantages and possible limitations of metal nanoparticle-based tumor theranostics, as well as the potential strategies to improve the in vivo performance of these unique materials.
Collapse
Affiliation(s)
- O A Kuchur
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - S A Tsymbal
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - M V Shestovskaya
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - N S Serov
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia
| | - M S Dukhinova
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia.
| | - A A Shtil
- International Institute 'Solution Chemistry of Advanced Materials and Technologies', ITMO University, 197101 Saint-Petersburg, Russia; Institute of Gene Biology, Russian Academy of Science, 119334 Moscow, Russia
| |
Collapse
|
8
|
Bolandparvaz A, Vapniarsky N, Harriman R, Alvarez K, Saini J, Zang Z, Van De Water J, Lewis JS. Biodistribution and toxicity of epitope-functionalized dextran iron oxide nanoparticles in a pregnant murine model. J Biomed Mater Res A 2020; 108:1186-1202. [PMID: 32031743 DOI: 10.1002/jbm.a.36893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
In pursuit of a preventive therapeutic for maternal autoantibody-related (MAR) autism, we assessed the toxicity, biodistribution, and clearance of a MAR specific peptide-functionalized dextran iron oxide nanoparticle system in pregnant murine dams. We previously synthesized ~15 nm citrate-coated dextran iron oxide nanoparticles (DIONPs), surface-modified with polyethylene glycol and MAR peptides to produce systems for nanoparticle-based autoantibody reception and entrapments (SNAREs). First, we investigated their immunogenicity and MAR lactate dehydrogenase B antibody uptake in murine serum in vitro. To assess biodistribution and toxicity, as well as systemic effects, we performed in vivo clinical and post mortem pathological evaluations. We observed minimal production of inflammatory cytokines-interleukin 10 (IL-10) and IL-12 following in vitro exposure of macrophages to SNAREs. We established the maximum tolerated dose of SNAREs to be 150 mg/kg at which deposition of iron was evident in the liver and lungs by histology and magnetic resonance imaging but no concurrent evidence of liver toxicity or lung infarction was detected. Further, SNAREs exhibited slower clearance from the maternal blood in pregnant dams compared to DIONPs based on serum total iron concentration. These findings demonstrated that the SNAREs have a prolonged presence in the blood and are safe for use in pregnant mice as evidenced by no associated organ damage, failure, inflammation, and fetal mortality. Determination of the MTD dose sets the basis for future studies investigating the efficacy of our nanoparticle formulation in a MAR autism mouse model.
Collapse
Affiliation(s)
- Amir Bolandparvaz
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Natalia Vapniarsky
- Department of Pathology Microbiology and Immunology, University of California Davis, Davis, California, USA
| | - Rian Harriman
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Kenneth Alvarez
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Jasmeen Saini
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Zexi Zang
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Judy Van De Water
- M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders), University of California Davis, Davis, California, USA.,Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, California, USA
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| |
Collapse
|
9
|
Comparative Molecular Immunological Activity of Physiological Metal Oxide Nanoparticle and its Anticancer Peptide and RNA Complexes. NANOMATERIALS 2019; 9:nano9121670. [PMID: 31771091 PMCID: PMC6955775 DOI: 10.3390/nano9121670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022]
Abstract
Currently, there is a great interest in nanoparticle-based vaccine delivery. Recent studies suggest that nanoparticles when introduced into the biological milieu are not simply passive carriers but may also contribute immunological activity themselves or of their own accord. For example there is considerable interest in the biomedical applications of one of the physiologically-based inorganic metal oxide nanoparticle, zinc oxide (ZnO). Indeed zinc oxide (ZnO) NP are now recognized as a nanoscale chemotherapeutic or anticancer nanoparticle (ANP) and several recent reports suggest ZnO NP and/or its complexes with drug and RNA induce a potent antitumor response in immuno-competent mouse models. A variety of cell culture studies have shown that ZnO NP can induce cytokines such as IFN-γ, TNF-α, IL-2, and IL-12 which are known to regulate the tumor microenvironment. Much less work has been done on magnesium oxide (MgO), cobalt oxide (Co3O4), or nickel oxide (NiO); however, despite the fact that these physiologically-based metal oxide NP are reported to functionally load and assemble RNA and protein onto their surface and may thus also be of potential interest as nanovaccine platform. Here we initially compared in vitro immunogenicity of ZnO and Co3O4 NP and their effects on cancer-associated or tolerogenic cytokines. Based on these data we moved ZnO NP forward to testing in the ex vivo splenocyte assay relative to MgO and NiO NP and these data showed significant difference for flow cytometry sorted population for ZnO-NP, relative to NiO and MgO. These data suggesting both molecular and cellular immunogenic activity, a double-stranded anticancer RNA (ACR), polyinosinic:poly cytidylic acid (poly I:C) known to bind ZnO NP; when ZnO-poly I:C was injected into B16F10-BALB/C tumor significantly induced, IL-2 and IL-12 as shown by Cohen’s d test. LL37 is an anticancer peptide (ACP) currently in clinical trials as an intratumoral immuno-therapeutic agent against metastatic melanoma. LL37 is known to bind poly I:C where it is thought to compete for receptor binding on the surface of some immune cells, metastatic melanoma and lung cells. Molecular dynamic simulations revealed association of LL37 onto ZnO NP confirmed by gel shift assay. Thus using the well-characterized model human lung cancer model cell line (BEAS-2B), poly I:C RNA, LL37 peptide, or LL37-poly I:C complexes were loaded onto ZnO NP and delivered to BEAS-2B lung cells, and the effect on the main cancer regulating cytokine, IL-6 determined by ELISA. Surprisingly ZnO-LL37, but not ZnO-poly I:C or the more novel tricomplex (ZnO-LL37-poly I:C) significantly suppressed IL-6 by >98–99%. These data support the further evaluation of physiological metal oxide compositions, so-called physiometacomposite (PMC) materials and their formulation with anticancer peptide (ACP) and/or anticancer RNA (ACR) as a potential new class of immuno-therapeutic against melanoma and potentially lung carcinoma or other cancers.
Collapse
|
10
|
Hsiao YP, Huang CH, Lin YC, Jan TR. Systemic exposure to a single dose of ferucarbotran aggravates neuroinflammation in a murine model of experimental autoimmune encephalomyelitis. Int J Nanomedicine 2019; 14:1229-1240. [PMID: 30863056 PMCID: PMC6391144 DOI: 10.2147/ijn.s189327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Medicinal preparations of iron oxide nanoparticles (IONPs) have been used as MRI contrast agents for the diagnosis of hepatic tumors and the assessment of neuroinflammation and blood–brain barrier integrity. However, it remains mostly unclear whether exposure to IONPs affects neuroinflammation under disease conditions. The present study aims to investigate the impact of IONPs on autoimmune-mediated neuroinflammation using a murine model of experimental autoimmune encephalomyelitis (EAE) that mimics human multiple sclerosis. Methods Mice were either left untreated or immunized with myelin oligodendrocyte glyco-protein on day 0 followed by two injections of pertussis toxin for EAE induction. The EAE mice were intravenously administered with a single dose of the carboxydextran-coated IONPs, ferucarbotran (20 mg Fe/kg) and/or saline (as vehicle) on day 18. Symptoms of EAE were daily monitored until the mice were killed on day 30. Tissue sections of the brain and spinal cord were prepared for histopathological examinations. Iron deposition, neuron demyelination and inflammatory cell infiltration were examined using histochemical staining. The infiltration of microglial and T cells, and cytokine expression were examined by immunohistochemical staining and/or reverse transcription polymerase chain reaction (RT-PCR). Results Iron deposition was detected in both the brain and spinal cord of EAE mice 3 days post-ferucarbotran treatment. The clinical and pathological scores of EAE, percentage of myelin loss and infiltration of inflammatory cells into the spinal cord were significantly deteriorated in EAE mice treated with ferucarbotran. Furthermore, ferucarbotran treatment increased the number of CD3+, Iba-1+, IL-6+, Iba-1+TNF-α+ and CD3+IFN-γ+ cells in the spinal cord of EAE mice. Conclusion A single exposure to ferucarbotran exacerbated neuroinflammation and disease severity of EAE, which might be attributed to the enhanced activation of microglia and T cells. These results demonstrated that the pro-inflammatory effect of ferucarbotran on the central nervous system is closely associated with the deterioration of autoimmunity.
Collapse
Affiliation(s)
- Yai-Ping Hsiao
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| | - Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Yu-Chin Lin
- Department of Medicinal Botanicals and Health Applications, College of Biotechnology & Bioresources, Da-Yeh University, Changhua, Taiwan
| | - Tong-Rong Jan
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| |
Collapse
|
11
|
Iron oxide nanoparticles modulate heat shock proteins and organ specific markers expression in mice male accessory organs. Toxicol Appl Pharmacol 2017; 317:12-24. [DOI: 10.1016/j.taap.2017.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/12/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022]
|
12
|
Valdiglesias V, Fernández-Bertólez N, Kiliç G, Costa C, Costa S, Fraga S, Bessa MJ, Pásaro E, Teixeira JP, Laffon B. Are iron oxide nanoparticles safe? Current knowledge and future perspectives. J Trace Elem Med Biol 2016; 38:53-63. [PMID: 27056797 DOI: 10.1016/j.jtemb.2016.03.017] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Due to their unique physicochemical properties, including superparamagnetism, iron oxide nanoparticles (ION) have a number of interesting applications, especially in the biomedical field, that make them one of the most fascinating nanomaterials. They are used as contrast agents for magnetic resonance imaging, in targeted drug delivery, and for induced hyperthermia cancer treatments. Together with these valuable uses, concerns regarding the onset of unexpected adverse health effects following exposure have been also raised. Nevertheless, despite the numerous ION purposes being explored, currently available information on their potential toxicity is still scarce and controversial data have been reported. Although ION have traditionally been considered as biocompatible - mainly on the basis of viability tests results - influence of nanoparticle surface coating, size, or dose, and of other experimental factors such as treatment time or cell type, has been demonstrated to be important for ION in vitro toxicity manifestation. In vivo studies have shown distribution of ION to different tissues and organs, including brain after passing the blood-brain barrier; nevertheless results from acute toxicity, genotoxicity, immunotoxicity, neurotoxicity and reproductive toxicity investigations in different animal models do not provide a clear overview on ION safety yet, and epidemiological studies are almost inexistent. Much work has still to be done to fully understand how these nanomaterials interact with cellular systems and what, if any, potential adverse health consequences can derive from ION exposure.
Collapse
Affiliation(s)
- Vanessa Valdiglesias
- DICOMOSA Group, Department of Psychology, Area of Psychobiology, Universidade da Coruña, Edificio de Servicios Centrales de Investigación, Campus Elviña s/n, A Coruña 15071, Spain
| | - Natalia Fernández-Bertólez
- DICOMOSA Group, Department of Psychology, Area of Psychobiology, Universidade da Coruña, Edificio de Servicios Centrales de Investigación, Campus Elviña s/n, A Coruña 15071, Spain; Department of Cell and Molecular Biology, Universidade da Coruña, Facultad de Ciencias, Campus A Zapateira s/n, A Coruña 15071, Spain
| | - Gözde Kiliç
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Carla Costa
- Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, Porto 4000-055, Portugal; EPIUnit-Institute of Public Health, University of Porto, Rua das Taipas, 135, Porto 4050-600, Portugal
| | - Solange Costa
- Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, Porto 4000-055, Portugal; EPIUnit-Institute of Public Health, University of Porto, Rua das Taipas, 135, Porto 4050-600, Portugal
| | - Sonia Fraga
- Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, Porto 4000-055, Portugal; EPIUnit-Institute of Public Health, University of Porto, Rua das Taipas, 135, Porto 4050-600, Portugal
| | - Maria Joao Bessa
- Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, Porto 4000-055, Portugal; EPIUnit-Institute of Public Health, University of Porto, Rua das Taipas, 135, Porto 4050-600, Portugal
| | - Eduardo Pásaro
- DICOMOSA Group, Department of Psychology, Area of Psychobiology, Universidade da Coruña, Edificio de Servicios Centrales de Investigación, Campus Elviña s/n, A Coruña 15071, Spain
| | - João Paulo Teixeira
- Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, Porto 4000-055, Portugal; EPIUnit-Institute of Public Health, University of Porto, Rua das Taipas, 135, Porto 4050-600, Portugal
| | - Blanca Laffon
- DICOMOSA Group, Department of Psychology, Area of Psychobiology, Universidade da Coruña, Edificio de Servicios Centrales de Investigación, Campus Elviña s/n, A Coruña 15071, Spain.
| |
Collapse
|
13
|
Wang X, Wang S, Zhang Y. Advance of the application of nano-controlled release system in ophthalmic drug delivery. Drug Deliv 2015; 23:2897-2901. [PMID: 26635087 DOI: 10.3109/10717544.2015.1116025] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ocular prescription application of nanometer materials are mainly concentrated in controlled release systems. Due to the unique properties of nanometer materials such as higher bioavailability and less side effects, it has great advantages in carrying ocular drugs of eye diseases compared with the traditional dosing method. As a result, nano-controlled release system has good application prospect in eye diseases. At present, a variety of different types of nano-controlled release systems have been used to enhance the efficiency of the ocular drugs including nanomicelles, nanoparticles, nanosuspensions, liposomes and dendrimers. In this article, the research progress and the application of nano-controlled release system in ophthalmic drug delivery are reviewed.
Collapse
Affiliation(s)
- Xuanzhong Wang
- a Ophthalmology Department , The 2nd Teaching Hospital of Jilin University , Changchun , China
| | - Shurong Wang
- a Ophthalmology Department , The 2nd Teaching Hospital of Jilin University , Changchun , China
| | - Yan Zhang
- a Ophthalmology Department , The 2nd Teaching Hospital of Jilin University , Changchun , China
| |
Collapse
|