1
|
Aminyavari S, Afshari AR, Ahmadi SS, Kesharwani P, Sanati M, Sahebkar A. Unveiling the theranostic potential of SPIONs in Alzheimer's disease management. J Psychiatr Res 2024; 179:244-256. [PMID: 39321523 DOI: 10.1016/j.jpsychires.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/22/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Alzheimer's disease (AD) is a devastating kind of dementia that is becoming more common worldwide. Toxic amyloid-beta (Aβ) aggregates are the primary cause of AD onset and development. Superparamagnetic iron oxide nanoparticles (SPIONs) have received a lot of interest in AD therapy over the last decade because of their ability to redirect the Aβ fibrillation process and improve associated brain dysfunction. The potential diagnostic application of SPIONs in AD has dramatically increased this interest. Furthermore, surface-modified engineered SPIONs function as drug carriers to improve the efficacy of current therapies. Various preclinical and clinical studies on the role of SPIONs in AD pathology have produced encouraging results. However, due to their physicochemical properties (e.g., size, surface charge, and particle concentration) in the biological milieu, SPIONs may play the role of a preventive or accelerative agent in AD. Even though SPIONs are potential therapeutic and diagnostic options in AD, significant efforts are still needed to overcome the inconsistencies and safety concerns. This review evaluated the current understanding of how various SPIONs interact with AD models and explored the discrepancies in their efficacy and safety.
Collapse
Affiliation(s)
- Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Seyed Sajad Ahmadi
- Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Shurin MR, Kirichenko VA, Shurin GV, Lee D, Crane C, Kirichenko AV. Radiomodulating Properties of Superparamagnetic Iron Oxide Nanoparticle (SPION) Agent Ferumoxytol on Human Monocytes: Implications for MRI-Guided Liver Radiotherapy. Cancers (Basel) 2024; 16:1318. [PMID: 38610996 PMCID: PMC11011128 DOI: 10.3390/cancers16071318] [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: 02/19/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) have attracted great attention not only for therapeutic applications but also as an alternative magnetic resonance imaging (MRI) contrast agent that helps visualize liver tumors during MRI-guided stereotactic body radiotherapy (SBRT). SPION can provide functional imaging of liver parenchyma based upon its uptake by the hepatic resident macrophages or Kupffer cells with a relative enhancement of malignant tumors that lack Kupffer cells. However, the radiomodulating properties of SPION on liver macrophages are not known. Utilizing human monocytic THP-1 undifferentiated and differentiated cells, we characterized the effect of ferumoxytol (Feraheme®), a carbohydrate-coated ultrasmall SPION agent at clinically relevant concentration and therapeutically relevant doses of gamma radiation on cultured cells in vitro. We showed that ferumoxytol affected both monocytes and macrophages, increased the resistance of monocytes to radiation-induced cell death and inhibition of cell activity, and supported the anti-inflammatory phenotype of human macrophages under radiation. Its effect on human cells depended on the duration of SPION uptake and was radiation dose-dependent. The results of this pilot study support a strong mechanism-based optimization of SPION-enhanced MRI-guided liver SBRT for primary and metastatic liver tumors, especially in patients with liver cirrhosis awaiting a liver transplant.
Collapse
Affiliation(s)
- Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
| | - Vladimir A. Kirichenko
- Department of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.A.K.); (D.L.)
| | - Galina V. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
| | - Danny Lee
- Department of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.A.K.); (D.L.)
| | - Christopher Crane
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Alexander V. Kirichenko
- Department of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.A.K.); (D.L.)
| |
Collapse
|
3
|
Pal S, de la Fuente IF, Sawant SS, Cannata JN, He W, Rouge JL. Cellular Uptake Mechanism of Nucleic Acid Nanocapsules and Their DNA-Surfactant Building Blocks. Bioconjug Chem 2023; 34:1004-1013. [PMID: 37231780 PMCID: PMC10330902 DOI: 10.1021/acs.bioconjchem.3c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nucleic acid nanocapsules (NANs) are enzyme-responsive DNA-functionalized micelles built for the controlled release of DNA-surfactant conjugates (DSCs) that present sequences with demonstrated therapeutic potential. Here, we investigate the mechanisms by which DSCs gain access to intracellular space in vitro and determine the effects of serum on the overall uptake and internalization mechanism of NANs. Using pharmacological inhibitors to selectively block certain pathways, we show, through confocal visualization of cellular distribution and flow cytometry quantification of total cellular association, that scavenger receptor-mediated, caveolae-dependent endocytosis is the major cellular uptake pathway of NANs in the presence and absence of serum. Furthermore, as NANs can be triggered to release DSCs by external stimuli such as enzymes, we sought to examine the uptake profile of particles degraded by enzymes prior to cell-based assays. We found that while scavenger receptor-mediated, caveolae-dependent endocytosis is still at play, energy-independent pathways as well as clathrin-mediated endocytosis are also involved. Overall, this study has helped to elucidate early steps in the cytosolic delivery and therapeutic activity of DSCs packaged into a micellular NAN platform while shedding light on the way in which DNA functionalized nanomaterials in general can be trafficked into cells both as nanostructures and as molecular entities. Importantly, our study also shows that the NAN design in particular is able to stabilize nucleic acids when delivered in the presence of serum, a critical step for effective therapeutic nucleic acid delivery.
Collapse
Affiliation(s)
- Suman Pal
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ina F de la Fuente
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Shraddha S Sawant
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jenna N Cannata
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Wu He
- Flow Cytometry Facility, Center for Open Research Resources and Equipment, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jessica L Rouge
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
4
|
Effects of Magnetic Nanoparticles on the Functional Activity of Human Monocytes and Dendritic Cells. Int J Mol Sci 2023; 24:ijms24021358. [PMID: 36674876 PMCID: PMC9864373 DOI: 10.3390/ijms24021358] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
The use of nanoparticles in medicine is sometimes hampered by their potential to activate immune cells, eliciting inflammation or allergy. We investigated whether magnetic nanoparticles (MNPs) or biomimetic magnetic nanoparticles (BMNPs) affect relevant activities of human monocytes. We found that the nanoparticles neither elicited the production of pro-inflammatory mediators IL-6 and TNFα by resting monocytes (when BMNP dose < 300 μg/mL) nor enhanced their secretion induced by R848, a molecule engaging virus-recognizing receptors, or bacterial lipopolysaccharide (LPS). MNPs and BMNPs neither induced the generation of reactive oxygen species (ROS), nor affected the ROS production elicited by the NADPH oxidase activator phorbol myristate acetate (PMA) or the fungal derivative β-glucan. BMNPs, but not MNPs, caused an up-regulation of the maturation markers CD80, CD83, and CD86 in immature monocyte-derived dendritic cells (DCs), whereas both nanoparticles did not affect the LPS-induced expression of these markers. Moreover, the nanoparticles were greedily ingested by monocytes and DCs without altering their viability. Therefore, these nanoparticles are candidates for medical applications because they do not activate pro-inflammatory activities of monocytes. Furthermore, their ability to stimulate DC maturation could be used for the design of vaccines. Moreover, harmlessly engulfed nanoparticles could be vehicles to carry molecules inside the immune cells to regulate the immune response.
Collapse
|
5
|
Chen Y, He X, Chen Q, He Y, Chen F, Yang C, Wang L. Nanomaterials against intracellular bacterial infection: from drug delivery to intrinsic biofunction. Front Bioeng Biotechnol 2023; 11:1197974. [PMID: 37180049 PMCID: PMC10174311 DOI: 10.3389/fbioe.2023.1197974] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Fighting intracellular bacteria with strong antibiotics evading remains a long-standing challenge. Responding to and regulating the infectious microenvironment is crucial for treating intracellular infections. Sophisticated nanomaterials with unique physicochemical properties exhibit great potential for precise drug delivery towards infection sites, along with modulating infectious microenvironment via their instinct bioactivity. In this review, we first identify the key characters and therapeutic targets of intracellular infection microenvironment. Next, we illustrate how the nanomaterials physicochemical properties, such as size, charge, shape and functionalization affect the interaction between nanomaterials, cells and bacteria. We also introduce the recent progress of nanomaterial-based targeted delivery and controlled release of antibiotics in intracellular infection microenvironment. Notably, we highlight the nanomaterials with unique intrinsic properties, such as metal toxicity and enzyme-like activity for the treatment of intracellular bacteria. Finally, we discuss the opportunities and challenges of bioactive nanomaterials in addressing intracellular infections.
Collapse
Affiliation(s)
- Yinglu Chen
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xiaoheng He
- Department of Applied Chemistry, Xi’an University of Technology, Xi’an, China
| | - Qiuhong Chen
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Fangman Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Macau SAR, China
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Liang Wang, ; Chao Yang,
| | - Liang Wang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Liang Wang, ; Chao Yang,
| |
Collapse
|
6
|
Functionalized magnetic nanoparticles for treating bone diseases. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
7
|
Kokhanyuk B, Vántus VB, Radnai B, Vámos E, Kajner G, Galbács G, Telek E, Mészáros M, Deli MA, Németh P, Engelmann P. Distinct Uptake Routes Participate in Silver Nanoparticle Engulfment by Earthworm and Human Immune Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2818. [PMID: 36014683 PMCID: PMC9413649 DOI: 10.3390/nano12162818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
The consequences of engineered silver nanoparticle (AgNP) exposure and cellular interaction with the immune system are poorly understood. The immunocytes of the Eisenia andrei earthworm are frequently applied in ecotoxicological studies and possess functional similarity to vertebrate macrophages. Hence, we characterized and compared the endocytosis mechanisms for the uptake of 75 nm AgNPs by earthworm coelomocytes, human THP-1 monocytes, and differentiated THP-1 (macrophage-like) cells. Our results indicate that microtubule-dependent, scavenger-receptor, and PI3K signaling-mediated macropinocytosis are utilized during AgNP engulfment by human THP-1 and differentiated THP-1 cells. However, earthworm coelomocytes employ actin-dependent phagocytosis during AgNPs uptake. In both human and earthworm immunocytes, AgNPs were located in the cytoplasm, within the endo-/lysosomes. We detected that the internalization of AgNPs is TLR/MyD88-dependent, also involving the bactericidal/permeability-increasing protein (BPI) in the case of human immunocytes. The exposure led to decreased mitochondrial respiration in human immunocytes; however, in coelomocytes, it enhanced respiratory parameters. Our findings provide more data about NP trafficking as nano-carriers in the nanomedicine field, as well as contribute to an understanding of the ecotoxicological consequences of nanoparticle exposure.
Collapse
Affiliation(s)
- Bohdana Kokhanyuk
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Viola Bagóné Vántus
- Department of Biochemistry and Medicinal Chemistry, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Balázs Radnai
- Department of Biochemistry and Medicinal Chemistry, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Eszter Vámos
- Department of Biochemistry and Medicinal Chemistry, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Gyula Kajner
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, H-6720 Szeged, Hungary
| | - Gábor Galbács
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, H-6720 Szeged, Hungary
| | - Elek Telek
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Péter Engelmann
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
8
|
Lu CH, Hsiao JK. Diagnostic and therapeutic roles of iron oxide nanoparticles in biomedicine. Tzu Chi Med J 2022; 35:11-17. [PMID: 36866343 PMCID: PMC9972926 DOI: 10.4103/tcmj.tcmj_65_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 06/08/2022] [Indexed: 11/04/2022] Open
Abstract
Nanotechnology changed our understanding of physics and chemics and influenced the biomedical field. Iron oxide nanoparticles (IONs) are one of the first emerging biomedical applications of nanotechnology. The IONs are composed of iron oxide core exhibiting magnetism and coated with biocompatible molecules. The small size, strong magnetism, and biocompatibility of IONs facilitate the application of IONs in the medical imaging field. We listed several clinical available IONs including Resovist (Bayer Schering Pharma, Berlin, Germany) and Feridex intravenous (I.V.)/Endorem as magnetic resonance (MR) contrast agents for liver tumor detection. We also illustrated GastroMARK as a gastrointestinal contrast agent for MR imaging. Recently, IONs named Feraheme for treating iron-deficiency anemia have been approved by the Food and Drug Administration. Moreover, tumor ablation by IONs named NanoTherm has also been discussed. In addition to the clinical application, several potential biomedical applications of IONs including cancer-targeting capability by conjugating IONs with cancer-specific ligands, cell trafficking tools, or tumor ablation agents have also been discussed. With the growing awareness of nanotechnology, further application of IONs is still on the horizon that would shed light on biomedicine.
Collapse
Affiliation(s)
- Chia-Hung Lu
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Jong-Kia Hsiao, Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 289, Jianguo Road, Xindian District, New Taipei, Taiwan. E-mail:
| |
Collapse
|
9
|
Thangudu S, Huang EY, Su CH. Safe magnetic resonance imaging on biocompatible nanoformulations. Biomater Sci 2022; 10:5032-5053. [PMID: 35858468 DOI: 10.1039/d2bm00692h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Magnetic resonance imaging (MRI) holds promise for the early clinical diagnosis of various diseases, but most clinical MR techniques require the use of a contrast medium. Several nanomaterial (NM) mediated contrast agents (CAs) are widely used as T1- and T2-based MR contrast agents for clinical and non-clinical applications. Unfortunately, most NM-based CAs are toxic or non-biocompatible, restricting their practical/clinical applications. Therefore, the development of nontoxic and biocompatible CAs for clinical MRI diagnosis is highly desired. To this end, several biocompatible and biomimetic strategies have been developed to offer long blood circulation time, significant biocompatibility, in vivo biodistribution and high contrast ability for efficient imaging. However, detailed review reports on biocompatible NMs, specifically for MR imaging have not yet been summarized. Thus, in the present review we summarize various surface coating strategies (such as polymers, proteins, cell membranes, etc.) to achieve biocompatible NPs, providing a detailed discussion of advances and future prospects for safe MRI imaging.
Collapse
Affiliation(s)
- Suresh Thangudu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
| | - Eng-Yen Huang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan. .,Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
10
|
Mohanty A, Parida A, Raut RK, Behera RK. Ferritin: A Promising Nanoreactor and Nanocarrier for Bionanotechnology. ACS BIO & MED CHEM AU 2022; 2:258-281. [PMID: 37101573 PMCID: PMC10114856 DOI: 10.1021/acsbiomedchemau.2c00003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
The essence of bionanotechnology lies in the application of nanotechnology/nanomaterials to solve the biological problems. Quantum dots and nanoparticles hold potential biomedical applications, but their inherent problems such as low solubility and associated toxicity due to their interactions at nonspecific target sites is a major concern. The self-assembled, thermostable, ferritin protein nanocages possessing natural iron scavenging ability have emerged as a potential solution to all the above-mentioned problems by acting as nanoreactor and nanocarrier. Ferritins, the cellular iron repositories, are hollow, spherical, symmetric multimeric protein nanocages, which sequester the excess of free Fe(II) and synthesize iron biominerals (Fe2O3·H2O) inside their ∼5-8 nm central cavity. The electrostatics and dynamics of the pore residues not only drives the natural substrate Fe2+ inside ferritin nanocages but also uptakes a set of other metals ions/counterions during in vitro synthesis of nanomaterial. The current review aims to report the recent developments/understanding on ferritin structure (self-assembly, surface/pores electrostatics, metal ion binding sites) and chemistry occurring inside these supramolecular protein cages (protein mediated metal ion uptake and mineralization/nanoparticle formation) along with its surface modification to exploit them for various nanobiotechnological applications. Furthermore, a better understanding of ferritin self-assembly would be highly useful for optimizing the incorporation of nanomaterials via the disassembly/reassembly approach. Several studies have reported the successful engineering of these ferritin protein nanocages in order to utilize them as potential nanoreactor for synthesizing/incorporating nanoparticles and as nanocarrier for delivering imaging agents/drugs at cell specific target sites. Therefore, the combination of nanoscience (nanomaterials) and bioscience (ferritin protein) projects several benefits for various applications ranging from electronics to medicine.
Collapse
|
11
|
Ngo W, Ahmed S, Blackadar C, Bussin B, Ji Q, Mladjenovic SM, Sepahi Z, Chan WC. Why nanoparticles prefer liver macrophage cell uptake in vivo. Adv Drug Deliv Rev 2022; 185:114238. [PMID: 35367524 DOI: 10.1016/j.addr.2022.114238] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/26/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023]
Abstract
Effective delivery of therapeutic and diagnostic nanoparticles is dependent on their ability to accumulate in diseased tissues. However, most nanoparticles end up in liver macrophages regardless of nanoparticle design after administration. In this review, we describe the interactions of liver macrophages with nanoparticles. Liver macrophages have significant advantages in interacting with circulating nanoparticles over most target cells and tissues in the body. We describe these advantages in this article. Understanding these advantages will enable the development of strategies to overcome liver macrophages and deliver nanoparticles to targeted diseased tissues effectively. Ultimately, these approaches will increase the therapeutic efficacy and diagnostic signal of nanoparticles.
Collapse
|
12
|
Prussian blue technique is prone to yield false negative results in magnetoreception research. Sci Rep 2022; 12:8803. [PMID: 35614116 PMCID: PMC9132912 DOI: 10.1038/s41598-022-12398-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/04/2022] [Indexed: 12/13/2022] Open
Abstract
Perls’s Prussian blue staining technique has been used in magnetoreception research to screen tissues for iron-rich structures as proxies for putative magnetoreceptor structures based on magnetic particles. However, seemingly promising structural candidates in the upper beak of birds detected with Prussian blue turned out to be either irreproducible or located in non-neuronal cells, which has spurred a controversy that has not been settled yet. Here we identify possible pitfalls in the previous works and apply the Prussian blue technique to tissues implicated in magnetic-particle-based magnetoreception, in an effort to reassess its suitability for staining single-domain magnetite, i.e., the proposed magnetic substrate for the interaction with the external magnetic field. In the upper beak of night-migratory songbirds, we found staining products in great numbers, but not remotely associated with fiber terminals of the traced ophthalmic branch of the trigeminal nerve. Surprisingly, staining products were absent from the lamina propria in the olfactory rosette of rainbow trout where candidate magnetoreceptor structures were identified with different techniques earlier. Critically, magnetosome chains in whole cells of magnetotactic bacteria remained unstained. The failure to label single-domain magnetite in positive control samples is a serious limitation of the technique and suggests that two most influential but antipodal studies conducted previously stood little chances of obtaining correct positive results under the assumption that magnetosome-like particles were present in the tissues. Nonetheless, the staining technique appears suitable to identify tissue contamination with iron-rich fine dust trapped in epithelia already in vivo.
Collapse
|
13
|
Application of Green Gold Nanoparticles in Cancer Therapy and Diagnosis. NANOMATERIALS 2022; 12:nano12071102. [PMID: 35407220 PMCID: PMC9000429 DOI: 10.3390/nano12071102] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023]
Abstract
Nanoparticles are currently used for cancer theranostics in the clinical field. Among nanoparticles, gold nanoparticles (AuNPs) attract much attention due to their usability and high performance in imaging techniques. The wide availability of biological precursors used in plant-based synthesized AuNPs allows for the development of large-scale production in a greener manner. Conventional cancer therapies, such as surgery and chemotherapy, have significant limitations and frequently fail to produce satisfying results. AuNPs have a prolonged circulation time, allow easy modification with ligands detected via cancer cell surface receptors, and increase uptake through receptor-mediated endocytosis. To exploit these unique features, studies have been carried out on the use of AuNPs as contrast agents for X-ray-based imaging techniques (i.e., computed tomography). As nanocarriers, AuNPs synthesized by nontoxic and biocompatible plants to deliver therapeutic biomolecules could be a significant stride forward in the effective treatment of various cancers. Fluorescent-plant-based markers, including AuNPs, fabricated using Medicago sativa, Olax Scandens, H. ambavilla, and H. lanceolatum, have been used in detecting cancers. Moreover, green synthesized AuNPs using various extracts have been applied for the treatment of different types of solid tumors. However, the cytotoxicity of AuNPs primarily depends on their size, surface reactivity, and surface area. In this review, the benefits of plant-based materials in cancer therapy are firstly explained. Then, considering the valuable position of AuNPs in medicine, the application of AuNPs in cancer therapy and detection is highlighted with an emphasis on limitations faced by the application of such NPs in drug delivery platforms.
Collapse
|
14
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
15
|
Jincy CS, Meena P. Evaluation of cytotoxic activity of Fe doped cobalt oxide nanoparticles. J Trace Elem Med Biol 2022; 70:126916. [PMID: 34995906 DOI: 10.1016/j.jtemb.2021.126916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/25/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND This contribution of work describes a new strategy for manufacturing cobalt oxide nanoparticles and the results assured that, its efficiency was increased by adding Fe ions. The anticancer drugs usually have a limited medical value owing to their nonspecific cytotoxicity. It has been proven that by using the nanosystems to deliver tablets to tumour cells reduces the toxic quality. Because of these qualities, nanoparticles can be used as a stronger rival for potent cancer treatment. METHOD This study investigated the cytotoxicity of iron doped cobalt oxide nanoparticles through trypan blue exclusion method. RESULT The newly generated Fe doped Co3O4 nanoparticles had proved its biocompatibility from the report of reduced toxicity below 200 μg/mL on malignant cell lines. CONCLUSION The observed findings may encourage the development of anticancer drugs based on the inorganic particles, especially Fe doped Co3O4 nanoparticles, that could be serve as an excellent framework for the drug delivery and provide a new perspective for interpreting and targeting various therapeutic methodologies to tumours.
Collapse
Affiliation(s)
- C S Jincy
- Department of Physics, PSGR Krishnammal College for Women, Peelamedu, Coimbatore, Tamilnadu, India.
| | - P Meena
- Associate Professor and Head, Department of Physics, PSGR Krishnammal College for Women, Peelamedu, Coimbatore, Tamilnadu, India.
| |
Collapse
|
16
|
Hepatic and renal cellular cytotoxic effects of heparin-coated superparamagnetic Iron oxide nanoparticles. Biomater Res 2021; 25:36. [PMID: 34736539 PMCID: PMC8567628 DOI: 10.1186/s40824-021-00241-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background Superparamagnetic iron oxide (SPIO) nanoparticles have been widely used in several biomedical engineering in vivo. Although various surface modifications have been made to these non-biodegradable nanoparticles to make them more biocompatible, their toxic potential still remains a major concern. Method In this study, we newly developed unfractionated heparin (UFH)-coated and low molecular weight heparin (LMWH)-coated SPIO nanoparticles through surface modification engineering, which was compared with commercially available dextran-coated SPIO nanoparticles. Their toxicity such as cytotoxicity, single cell gel electrophoresis (SCGE) comet assay, intracellular reactive oxygen species (ROS) content and cellular apoptosis was evaluated to hepatic HepG2 and renal HK-2 cells. Results When UFH-, LMWH- or dextran-coated SPIO nanoparticles were applied, they did not affect the viability of HepG2 cell. However, HK-2 cells were more sensitive to dextran-coated SPIO nanoparticles than others. In genotoxicity assay using SCGE comet, DNA tail moment values in the groups treated with dextran- and LMWH-coated SPIO nanoparticles significantly increased. However, UFH-coated SPIO nanoparticles was only significantly lowing DNA tail moment value. In addition, UFH-coated SPIO nanoparticles had lower cytotoxicity in HepG2 and HK-2 cells compared to dextran-coated SPIO nanoparticles, especially in terms of apoptosis and intracellular ROS production. Conclusions Collectively, it is possible that UFH- coated SPIO nanoparticles can be used as alternative negative contrast agents.
Collapse
|
17
|
Della Camera G, Madej M, Ferretti AM, La Spina R, Li Y, Corteggio A, Heinzl T, Swartzwelter BJ, Sipos G, Gioria S, Ponti A, Boraschi D, Italiani P. Personalised Profiling of Innate Immune Memory Induced by Nano-Imaging Particles in Human Monocytes. Front Immunol 2021; 12:692165. [PMID: 34421901 PMCID: PMC8377278 DOI: 10.3389/fimmu.2021.692165] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022] Open
Abstract
Engineered nanoparticles used for medical purposes must meet stringent safety criteria, which include immunosafety, i.e., the inability to activate possibly detrimental immune/inflammatory effects. Even medical nanomaterials devoid of direct immunotoxic or inflammatory effects may have an impact on human health if able to modify innate memory, which is the ability to “prime” future immune responses towards a different, possibly more detrimental reactivity. Although innate memory is usually protective, anomalous innate memory responses may be at the basis of immune pathologies. In this study, we have examined the ability of two nanomaterials commonly used for diagnostic imaging purposes, gold and iron oxide nanoparticles, to induce or modulate innate memory, using an in vitro model based on human primary monocytes. Monocytes were exposed in culture to nanoparticles alone or together with the bacterial agent LPS (priming phase/primary response), then rested for six days (extinction phase), and eventually challenged with LPS (memory/secondary response). The memory response to the LPS challenge was measured as changes in the production of inflammatory (TNFα, IL-6) and anti-inflammatory cytokines (IL-10, IL-1Ra), as compared to unprimed monocytes. The results show that both types of nanoparticles can have an effect in the induction of memory, with changes observed in the cytokine production. By comparing nanomaterials of different shapes (spherical vs. rod-shaped gold particles) and different size (17 vs. 22 nm diameter spherical iron oxide particles), it was evident that innate memory could be differentially induced and modulated depending on size, shape and chemical composition. However, the main finding was that the innate memory effect of the particles was strongly donor-dependent, with monocytes from each donor showing a distinct memory profile upon priming with the same particles, thereby making impossible to draw general conclusions on the particle effects. Thus, in order to predict the effect of imaging nanoparticles on the innate memory of patients, a personalised profiling would be required, able to take in consideration the peculiarities of the individual innate immune reactivity.
Collapse
Affiliation(s)
- Giacomo Della Camera
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Mariusz Madej
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Anna Maria Ferretti
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), National Research Council (CNR), Milano, Italy
| | - Rita La Spina
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Yang Li
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Annunziata Corteggio
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Tommaso Heinzl
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Benjamin J Swartzwelter
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Gergö Sipos
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Alessandro Ponti
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), National Research Council (CNR), Milano, Italy
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy.,Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| |
Collapse
|
18
|
Chauhan DS, Dhasmana A, Laskar P, Prasad R, Jain NK, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Nanotechnology synergized immunoengineering for cancer. Eur J Pharm Biopharm 2021; 163:72-101. [PMID: 33774162 PMCID: PMC8170847 DOI: 10.1016/j.ejpb.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022]
Abstract
Novel strategies modulating the immune system yielded enhanced anticancer responses and improved cancer survival. Nevertheless, the success rate of immunotherapy in cancer treatment has been below expectation(s) due to unpredictable efficacy and off-target effects from systemic dosing of immunotherapeutic(s). As a result, there is an unmet clinical need for improving conventional immunotherapy. Nanotechnology offers several new strategies, multimodality, and multiplex biological targeting advantage to overcome many of these challenges. These efforts enable programming the pharmacodynamics, pharmacokinetics, and delivery of immunomodulatory agents/co-delivery of compounds to prime at the tumor sites for improved therapeutic benefits. This review provides an overview of the design and clinical principles of biomaterials driven nanotechnology and their potential use in personalized nanomedicines, vaccines, localized tumor modulation, and delivery strategies for cancer immunotherapy. In this review, we also summarize the latest highlights and recent advances in combinatorial therapies availed in the treatment of cold and complicated tumors. It also presents key steps and parameters implemented for clinical success. Finally, we analyse, discuss, and provide clinical perspectives on the integrated opportunities of nanotechnology and immunology to achieve synergistic and durable responses in cancer treatment.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nishant K Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
19
|
Ghanbarei S, Sattarahmady N, Zarghampoor F, Azarpira N, Hossein-Aghdaie M. Effects of labeling human mesenchymal stem cells with superparamagnetic zinc-nickel ferrite nanoparticles on cellular characteristics and adipogenesis/osteogenesis differentiation. Biotechnol Lett 2021; 43:1659-1673. [PMID: 33934256 DOI: 10.1007/s10529-021-03134-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/15/2021] [Indexed: 12/28/2022]
Abstract
OBJECTIVE An attractive cell source for stem cell-based therapy are WJ-MSCs. Hence, tracking WJ-MSCs using non-invasive imaging procedures (such as MRI) and contrast agents (Zn0.5Ni0.5Fe2O4, NFNPs) are required to evaluate cell distribution, migration, and differentiation. RESULTS Results showed that the bare and dextrin-coated NFNPs were internalized inside the WJ-MSCs and had no effect on the cell viability, proliferation, apoptosis, karyotyping, and morphology of WJ-MSCs up to 125 µg/mL. Besides, treated WJ-MSCs were differentiated into osteo/adipocyte-like cells. The expression of RUNX 2, SPP 1 (P < 0.05), and OCN (P > 0.05) genes in the WJ-MSCs treated with dextrin-coated NFNPs was higher than the untreated WJ-MSCs; and the expression of CFD, LPL, and PPAR-γ genes was reduced in WJ-MSCs treated with both NFNPs in comparison with the untreated WJ-MSCs (P > 0.05). CONCLUSION Overall, results showed that dextrin-coated NFNPs had no adverse effect on the cellular characteristics, proliferation, and differentiation of WJ-MSCs, and suggesting their potential clinical efficacy.
Collapse
Affiliation(s)
- Solaleh Ghanbarei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran.,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Naghmeh Sattarahmady
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,The Nanobiology and Nanomedicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Zarghampoor
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Khalili St, Mohamad Rasoolalah Research Tower, 7th floor, Shiraz, Iran.
| | - Negar Azarpira
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Khalili St, Mohamad Rasoolalah Research Tower, 7th floor, Shiraz, Iran.
| | | |
Collapse
|
20
|
Klein S, Distel LVR, Neuhuber W, Kryschi C. Caffeic Acid, Quercetin and 5-Fluorocytidine-Functionalized Au-Fe 3O 4 Nanoheterodimers for X-ray-Triggered Drug Delivery in Breast Tumor Spheroids. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1167. [PMID: 33947086 PMCID: PMC8146450 DOI: 10.3390/nano11051167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023]
Abstract
Au-Fe3O4 nanoheterodimers (NHD) were functionalized with the natural and synthetic anticancer drugs caffeic acid (CA), quercetin (Q) and 5-fluorocytidine (5FC). Their X-radiation dose-enhancing potential and chemotherapeutic efficacy for bimodal cancer therapy were investigated by designing multicellular tumor spheroids (MCTS) to in vitro avascular tumor models. MCTS were grown from the breast cancer cell lines MCF-7, MDA-MB-231, and MCF-10A. The MCF-7, MDA-MB-231 and MCF-10A MCTS were incubated with NHD-CA, NHD-Q, or NHD-5FC and then exposed to fractionated X-radiation comprising either a single 10 Gy dose, 2 daily single 5 Gy doses or 5 daily single 2 Gy doses. The NHD-CA, NHD-Q, and NHD-5FC affected the growth of X-ray irradiated and non-irradiated MCTS in a different manner. The impact of the NHDs on the glycolytic metabolism due to oxygen deprivation inside MCTS was assessed by measuring lactate secretion and glucose uptake by the MCTS. The NHD-CA and NHD-Q were found to act as X-radiation dose agents in MCF-7 MCTS and MDA-MB-231 MCTS and served as radioprotector in MCF-10A MCTS. X-ray triggered release of CA and Q inhibited lactate secretion and thereupon disturbed glycolytic reprogramming, whereas 5FC exerted their cytotoxic effects on both, healthy and tumor cells, after their release into the cytosol.
Collapse
Affiliation(s)
- Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr. 3, D-91058 Erlangen, Germany;
| | - Luitpold V. R. Distel
- Department of Radiation Oncology, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, D-91054 Erlangen, Germany;
| | - Winfried Neuhuber
- Institute of Anatomy, Chair of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nuremberg, Krankenhausstr. 9, D-91054 Erlangen, Germany;
| | - Carola Kryschi
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr. 3, D-91058 Erlangen, Germany;
| |
Collapse
|
21
|
Lorkowski ME, Atukorale PU, Ghaghada KB, Karathanasis E. Stimuli-Responsive Iron Oxide Nanotheranostics: A Versatile and Powerful Approach for Cancer Therapy. Adv Healthc Mater 2021; 10:e2001044. [PMID: 33225633 PMCID: PMC7933107 DOI: 10.1002/adhm.202001044] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/14/2020] [Indexed: 12/16/2022]
Abstract
Recent advancements in unravelling elements of cancer biology involved in disease progression and treatment resistance have highlighted the need for a holistic approach to effectively tackle cancer. Stimuli-responsive nanotheranostics based on iron oxide nanoparticles are an emerging class of versatile nanomedicines with powerful capabilities to "seek, sense, and attack" multiple components of solid tumors. In this work, the rationale for using iron oxide nanoparticles and the basic physical principles that impact their function in biomedical applications are reviewed. Subsequently, recent advances in the integration of iron oxide nanoparticles with various stimulus mechanisms to facilitate the development of stimuli-responsive nanotheranostics for application in cancer therapy are summarized. The integration of an iron oxide core with various surface coating mechanisms results in the generation of hybrid nanoconstructs with capabilities to codeliver a wide variety of highly potent anticancer therapeutics and immune modulators. Finally, emerging future directions and considerations for their clinical translation are touched upon.
Collapse
Affiliation(s)
- Morgan E. Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Prabhani U. Atukorale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ketan B. Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas, USA
- Department of Radiology, Baylor College of Medicine, Houston, Texas, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
22
|
He Y, Yi C, Zhang X, Zhao W, Yu D. Magnetic graphene oxide: Synthesis approaches, physicochemical characteristics, and biomedical applications. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116191] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Portilla Y, Mellid S, Paradela A, Ramos-Fernández A, Daviu N, Sanz-Ortega L, Pérez-Yagüe S, Morales MP, Barber DF. Iron Oxide Nanoparticle Coatings Dictate Cell Outcomes Despite the Influence of Protein Coronas. ACS APPLIED MATERIALS & INTERFACES 2021; 13:7924-7944. [PMID: 33587585 DOI: 10.1021/acsami.0c20066] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A critical issue in nanomedicine is to understand the complex dynamics that dictate the interactions of nanoparticles (NPs) with their biological milieu. The most exposed part of a nanoparticle is its surface coating, which comes into contact with the biological medium and adsorbs proteins, forming what is known as a protein corona (PC). It is assumed that this PC mainly dictates the nanoparticle-cell interactions. As such, we set out to analyze how different coatings on iron oxide nanoparticles (MNPs) affect the composition of the PC that forms on top of them, and how these newly formed coronas influence the uptake of MNPs by macrophages and tumor cells, their subcellular location upon internalization, and their intracellular degradation. We found that different superficial charges of the coatings did not affect the PC composition, with an enrichment in proteins with affinity for divalent ions regardless of the type of coating. The iron oxide core of the MNP might become exposed to the biological medium, influencing the proteins that constitute the PCs. The presence of enzymes with hydrolase activity in the PC could explain the degradation of the coatings when they come into contact with the biological media. In terms of MNP internalization by cells, coatings mainly determine the endocytic pathways used, especially in terms of receptor-mediated endocytosis. However, the increase in hydrodynamic size provoked by the formation of the associated corona drives uptake mechanisms like macropinocytosis. Once inside the cells, the PC protected the NPs in their intracellular transit to lysosomes, where they were fully degraded. This understanding of how coatings and PCs influence different cellular processes will help design improved NPs for biomedical applications, taking into account the influence of the coating and corona on the biology of the NPs.
Collapse
Affiliation(s)
- Yadileiny Portilla
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Sara Mellid
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Alberto Paradela
- Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Antonio Ramos-Fernández
- Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Neus Daviu
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Laura Sanz-Ortega
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - María P Morales
- Department of Energy, Environment and Health, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| |
Collapse
|
24
|
Sun X, Wang Y, Wen S, Huang K, Huang J, Chu X, Wang F, Pang L. Novel controlled and targeted releasing hydrogen sulfide system exerts combinational cerebral and myocardial protection after cardiac arrest. J Nanobiotechnology 2021; 19:40. [PMID: 33549092 PMCID: PMC7866762 DOI: 10.1186/s12951-021-00784-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cardiac arrest (CA) is a leading cause of death worldwide. Even after successful cardiopulmonary resuscitation (CPR), the majorities of survivals are companied with permanent myocardial and cerebral injury. Hydrogen sulfide (H2S) has been recognized as a novel gasotransmitter exerting multiple organ protection; however, the lacks of ideal H2S donors which can controlled release H2S to targeted organs such as heart and brain limits its application. RESULTS This work utilized mesoporous iron oxide nanoparticle (MION) as the carriers of diallyl trisulfide (DATS), with polyethylene glycol (PEG) and lactoferrin (LF) modified to MIONs to acquire the prolonged circulation time and brain-targeting effects, and a novel targeted H2S releasing system was constructed (DATS@MION-PEG-LF), which exhibited excellent biocompatibility, controlled-releasing H2S pattern, heart and brain targeting features, and the ability to be non-invasive traced by magnetic resonance imaging. DATS@MION-PEG-LF presented potent protective effects against cerebral and cardiac ischemic injury after CA in both in vitro hypoxia/reoxygenation models and in vivo CA/CPR models, which mainly involves anti-apoptosis, anti-inflammatory and anti-oxidant mechanisms. Accordingly, the cardiac and cerebral functions were obviously improved after CA/CPR, with potentially improved survival. CONCLUSIONS The present work provides a unique platform for targeted controlled release of H2S based on MIONs, and offers a new method for combinational myocardial and cerebral protection from ischemic injury, bringing considerable benefits for CA patients.
Collapse
Affiliation(s)
- Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China.
| | - Yiqing Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Shuyan Wen
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Kai Huang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Jiechun Huang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Xianglin Chu
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Fangrui Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| | - Liewen Pang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, 12th Wulumuqi Rd, 200040, Shanghai, China
| |
Collapse
|
25
|
Abstract
It is known that iron is found as a trace element in bone tissue, the main inorganic constituent of which is hydroxyapatite. Therefore, iron-doped hydroxyapatite (HApFe) materials could be new alternatives for many biomedical applications. A facile dip coating process was used to elaborate the iron-doped hydroxyapatite (HApFe) nanocomposite coatings. The HApFe suspension used to prepare the coatings was achieved using a co-precipitation method, which was adapted in the laboratory. The quality of the HApFe suspension was assessed through dynamic light scattering (DLS), ultrasonic measurements, and zeta potential values. The hydroxyapatite XRD patterns were observed in the HApFe nanocomposite with no significant shifting of peak positions, thus suggesting that the incorporation of iron did not significantly modify the hydroxyapatite structure. The morphology of the HApFe nanoparticles was evaluated using transmission electron microscopy (TEM). Scanning electron microscopy (SEM) was used in order to investigate the morphologies of HApFe particles and coatings, while their chemical compositions were assessed using energy-dispersive X-ray spectroscopy (EDS). The SEM results suggested that the HApFe consists mainly of spherical nanometric particles and that the surfaces of the coatings are continuous and homogeneous. Additionally, the EDS spectra highlighted the purity of the samples and confirmed the presence of calcium, phosphorous, and iron in the analyzed sample. The in vitro cytotoxicity of the HApFe suspensions and coatings was evidenced using osteoblast cells. The MTT assay showed that both the HApFe suspensions and coatings exhibited biocompatible properties.
Collapse
|
26
|
Leitner NS, Schroffenegger M, Reimhult E. Polymer Brush-Grafted Nanoparticles Preferentially Interact with Opsonins and Albumin. ACS APPLIED BIO MATERIALS 2020; 4:795-806. [PMID: 33490885 PMCID: PMC7818653 DOI: 10.1021/acsabm.0c01355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
![]()
Nanoparticles
find increasing applications in life science and
biomedicine. The fate of nanoparticles in a biological system is determined
by their protein corona, as remodeling of their surface properties
through protein adsorption triggers specific recognition such as cell
uptake and immune system clearance and nonspecific processes such
as aggregation and precipitation. The corona is a result of nanoparticle–protein
and protein–protein interactions and is influenced by particle
design. The state-of-the-art design of biomedical nanoparticles is
the core–shell structure exemplified by superparamagnetic iron
oxide nanoparticles (SPIONs) grafted with dense, well-hydrated polymer
shells used for biomedical magnetic imaging and therapy. Densely grafted
polymer chains form a polymer brush, yielding a highly repulsive barrier
to the formation of a protein corona via nonspecific
particle–protein interactions. However, recent studies showed
that the abundant blood serum protein albumin interacts with dense
polymer brush-grafted SPIONs. Herein, we use isothermal titration
calorimetry to characterize the nonspecific interactions between human
serum albumin, human serum immunoglobulin G, human transferrin, and
hen egg lysozyme with monodisperse poly(2-alkyl-2-oxazoline)-grafted
SPIONs with different grafting densities and core sizes. These particles
show similar protein interactions despite their different “stealth”
capabilities in cell culture. The SPIONs resist attractive interactions
with lysozymes and transferrins, but they both show a significant
exothermic enthalpic and low exothermic entropic interaction with
low stoichiometry for albumin and immunoglobulin G. Our results highlight
that protein size, flexibility, and charge are important to predict
protein corona formation on polymer brush-stabilized nanoparticles.
Collapse
Affiliation(s)
- Nikolaus Simon Leitner
- Institute for Biologically Inspired Materials, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Vienna A-1190, Vienna, Austria
| | - Martina Schroffenegger
- Institute for Biologically Inspired Materials, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Vienna A-1190, Vienna, Austria
| | - Erik Reimhult
- Institute for Biologically Inspired Materials, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Vienna A-1190, Vienna, Austria
| |
Collapse
|
27
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Kubinová Š, Dejneka A, Lunov O. Analyzing the mechanisms of iron oxide nanoparticles interactions with cells: A road from failure to success in clinical applications. J Control Release 2020; 328:59-77. [DOI: 10.1016/j.jconrel.2020.08.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022]
|
28
|
Vakili-Ghartavol R, Momtazi-Borojeni AA, Vakili-Ghartavol Z, Aiyelabegan HT, Jaafari MR, Rezayat SM, Arbabi Bidgoli S. Toxicity assessment of superparamagnetic iron oxide nanoparticles in different tissues. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:443-451. [PMID: 32024389 DOI: 10.1080/21691401.2019.1709855] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been employed in several biomedical applications where they facilitate both diagnostic and therapeutic aims. Although the potential benefits of SPIONs with different surface chemistry and conjugated targeting ligands/proteins are considerable, complicated interactions between these nanoparticles (NPs) and cells leading to toxic impacts could limit their clinical applications. Hence, elevation of our knowledge regarding the SPION-related toxicity is necessary. Here, the present review article will consider current studies and compare the potential toxic effect of SPIONs with or without identical surface chemistries on different cell lines. It centers on cellular and molecular mechanisms underlying toxicity of SPIONs. Likewise, emphasis is being dedicated for toxicity of SPIONs in various cell lines, in vitro and animal models, in vivo.
Collapse
Affiliation(s)
- Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Momtazi-Borojeni
- Halal Research Center of IRI, FDA, Tehran, Iran.,Nanotechnology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeynab Vakili-Ghartavol
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hammed Tanimowo Aiyelabegan
- Department of Medical Biochemistry and Pharmacology, College of Pure and Applied Sciences, Kwara State University Malete, Nigeria
| | - Mahmoud Reza Jaafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Sepideh Arbabi Bidgoli
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| |
Collapse
|
29
|
Nie Y, Rui Y, Miao C, Li Q, Hu F, Gu H. A stable USPIO capable for MR lymphography with ultra-low effective dosage. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102233. [PMID: 32522710 DOI: 10.1016/j.nano.2020.102233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/01/2023]
Abstract
Ultra-small superparamagnetic iron oxide (USPIO) nanoparticles appear to be promising tools for MR lymphography due to their unique magnetic properties. In clinical diagnosis, the effectiveness of USPIO will greatly affect the clinician's judgment to the enhanced MR images. In this study, we evaluated the effectiveness of CS015, a PAA-coated USPIO, with subcutaneous and intravenous administration. It appeared that subcutaneously injected particles had much higher efficiency to reach lymph nodes, and even worked at a very small dose 0.075 μmol/kg. Further, we compared CS015 with ferumoxytol and ferumoxtran-10 in MR lymphography and found that CS015 had the best performance. And the lymph node metastases in New Zealand rabbits were successfully detected using CS015 with one single dose. These merits of CS015 make it a promising MR lymphography contrast agent with potential applications in cancer therapy.
Collapse
Affiliation(s)
- Ying Nie
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanpeng Rui
- Department of Radiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chongchong Miao
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Qinshan Li
- So-Fe Biomedicine, Xuhui District, Shanghai, China
| | - Fenglin Hu
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Hongchen Gu
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
30
|
The Intrinsic Biological Identities of Iron Oxide Nanoparticles and Their Coatings: Unexplored Territory for Combinatorial Therapies. NANOMATERIALS 2020; 10:nano10050837. [PMID: 32349362 PMCID: PMC7712800 DOI: 10.3390/nano10050837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Over the last 20 years, iron oxide nanoparticles (IONPs) have been the subject of increasing investigation due to their potential use as theranostic agents. Their unique physical properties (physical identity), ample possibilities for surface modifications (synthetic identity), and the complex dynamics of their interaction with biological systems (biological identity) make IONPs a unique and fruitful resource for developing magnetic field-based therapeutic and diagnostic approaches to the treatment of diseases such as cancer. Like all nanomaterials, IONPs also interact with different cell types in vivo, a characteristic that ultimately determines their activity over the short and long term. Cells of the mononuclear phagocytic system (macrophages), dendritic cells (DCs), and endothelial cells (ECs) are engaged in the bulk of IONP encounters in the organism, and also determine IONP biodistribution. Therefore, the biological effects that IONPs trigger in these cells (biological identity) are of utmost importance to better understand and refine the efficacy of IONP-based theranostics. In the present review, which is focused on anti-cancer therapy, we discuss recent findings on the biological identities of IONPs, particularly as concerns their interactions with myeloid, endothelial, and tumor cells. Furthermore, we thoroughly discuss current understandings of the basic molecular mechanisms and complex interactions that govern IONP biological identity, and how these traits could be used as a stepping stone for future research.
Collapse
|
31
|
Wang P, Kim T, Harada M, Contag C, Huang X, Smith BR. Nano-immunoimaging. NANOSCALE HORIZONS 2020; 5:628-653. [PMID: 32226975 DOI: 10.1039/c9nh00514e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunoimaging is a rapidly growing field stoked in large part by the intriguing triumphs of immunotherapy. On the heels of immunotherapy's successes, there exists a growing need to evaluate tumor response to therapy particularly immunotherapy, stratify patients into responders vs. non-responders, identify inflammation, and better understand the fundamental roles of immune system components to improve both immunoimaging and immunotherapy. Innovative nanomaterials have begun to provide novel opportunities for immunoimaging, in part due to their sensitivity, modularity, capacity for many potentially varied ligands (high avidity), and potential for multifunctionality/multimodality imaging. This review strives to comprehensively summarize the integration of nanotechnology and immunoimaging, and the field's potential for clinical applications.
Collapse
Affiliation(s)
- Ping Wang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA
| | - Taeho Kim
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 488824, USA
| | - Xuefei Huang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Chemistry, Michigan State University, East Lansing, MI 488824, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Radiology, Stanford University, Stanford, CA 94306, USA
| |
Collapse
|
32
|
Yusefi M, Shameli K, Ali RR, Pang SW, Teow SY. Evaluating Anticancer Activity of Plant-Mediated Synthesized Iron Oxide Nanoparticles Using Punica Granatum Fruit Peel Extract. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127539] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
Guggenheim EJ, Rappoport JZ, Lynch I. Mechanisms for cellular uptake of nanosized clinical MRI contrast agents. Nanotoxicology 2020; 14:504-532. [PMID: 32037933 DOI: 10.1080/17435390.2019.1698779] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Engineered Nanomaterials (NMs), such as Superparamagnetic Iron Oxide Nanoparticles (SPIONs), offer significant benefits in a wide range of applications, including cancer diagnostic and therapeutic strategies. However, the use of NMs in biomedicine raises safety concerns due to lack of knowledge on possible biological interactions and effects. The initial basis for using SPIONs as biomedical MRI contrast enhancement agents was the idea that they are selectively taken up by macrophage cells, and not by the surrounding cancer cells. To investigate this claim, we analyzed the uptake of SPIONs into well-established cancer cell models and benchmarked this against a common macrophage cell model. In combination with fluorescent labeling of compartments and siRNA silencing of various proteins involved in common endocytic pathways, the mechanisms of internalization of SPIONs in these cell types has been ascertained utilizing reflectance confocal microscopy. Caveolar mediated endocytosis and macropinocytosis are both implicated in SPION uptake into cancer cells, whereas in macrophage cells, a clathrin-dependant route appears to predominate. Colocalization studies confirmed the eventual fate of SPIONs as accumulation in the degradative lysosomes. Dissolution of the SPIONs within the lysosomal environment has also been determined, allowing a fuller understanding of the cellular interactions, uptake, trafficking and effects of SPIONs within a variety of cancer cells and macrophages. Overall, the behavior of SPIONS in non-phagocytotic cell lines is broadly similar to that in the specialist macrophage cells, although some differences in the uptake patterns are apparent.
Collapse
Affiliation(s)
- Emily J Guggenheim
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Joshua Z Rappoport
- Center for Advanced Microscopy, and Nikon Imaging Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Core Technologies for Life Sciences, Boston College, MA, USA
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
34
|
Liu L, Jin R, Duan J, Yang L, Cai Z, Zhu W, Nie Y, He J, Xia C, Gong Q, Song B, Anderson JM, Ai H. Bioactive iron oxide nanoparticles suppress osteoclastogenesis and ovariectomy-induced bone loss through regulating the TRAF6-p62-CYLD signaling complex. Acta Biomater 2020; 103:281-292. [PMID: 31866569 DOI: 10.1016/j.actbio.2019.12.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 02/05/2023]
Abstract
Iron oxide nanoparticles (IONPs) have been widely used as contrast agents for magnetic resonance imaging (MRI) and other biomedical applications in both clinical and preclinical cases. In the present study, we show that two clinically used IONPs, ferumoxytol and ferucarbotran, have an intrinsic inhibitory effect on receptor activator NF-κB ligand (RANKL)-induced osteoclastogenesis of bone marrow-derived monocytes/macrophages (BMMs). IONPs significantly inhibited the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts and functional actin ring structures. More importantly, the inhibitory effect was also verified in vivo by its capacity to rescue the bone loss of ovariectomized (OVX) mice after intravenous injection with IONPs. Mechanistically, we found that IONPs trigger the upregulation of p62 which result in recruitment of CYLD and enhanced deubiquitination of TRAF6, a master controller of RANKL signaling. The downstream activation of NF-κB and MAPK signals was accordingly attenuated, ultimately leading to reduced expression of osteoclatogenesis-related genes. Taken together, clinically used IONPs can inhibit osteoclastogenesis through regulating TRAF6-p62-CYLD signaling complex, and they may be considered as alternative options for treatment of osteoporosis. STATEMENT OF SIGNIFICANCE: Nanoparticles have been developed as drug delivery systems for treatment of osteoporosis, mostly an age-related health problem with risk of fractures. In this work, we show that two clinically used iron oxide nanoparticles (IONPs) ferumoxytol and ferucarbotran themselves can significantly reduce the osteoporosis of ovariectomized (OVX) mice through inhibiting Osteoclastogenesis. We found that IONPs trigger the upregulation of p62 which result in recruitment of CYLD and enhanced deubiquitination of TRAF6, a master controller of RANKL signaling. The downstream activation of NF-κB and MAPK signals was accordingly attenuated, leading to reduced expression of osteoclatogenesis-related genes. Taken together, clinically used IONPs inhibit osteoclastogenesis through regulating TRAF6-p62-CYLD signaling complex, and they may be considered as alternative options for treatment of osteoporosis.
Collapse
Affiliation(s)
- Li Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Jimei Duan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Wencheng Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - James M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
35
|
Sun Y, Shi F, Niu Y, Zhang Y, Xiong F. Fe3O4@OA@Poloxamer nanoparticles lower triglyceride in hepatocytes through liposuction effect and nano-enzyme effect. Colloids Surf B Biointerfaces 2019; 184:110528. [DOI: 10.1016/j.colsurfb.2019.110528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/06/2019] [Accepted: 09/24/2019] [Indexed: 01/09/2023]
|
36
|
Alghsham RS, Satpathy SR, Bodduluri SR, Hegde B, Jala VR, Twal W, Burlison JA, Sunkara M, Haribabu B. Zinc Oxide Nanowires Exposure Induces a Distinct Inflammatory Response via CCL11-Mediated Eosinophil Recruitment. Front Immunol 2019; 10:2604. [PMID: 31787980 PMCID: PMC6856074 DOI: 10.3389/fimmu.2019.02604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 01/21/2023] Open
Abstract
High aspect ratio zinc oxide nanowires (ZnONWs) have become one of the most important products in nanotechnology. The wide range applications of ZnONWs have heightened the need for evaluating the risks and biological consequences to these particles. In this study, we investigated inflammatory pathways activated by ZnONWs in cultured cells as well as the consequences of systemic exposure in mouse models. Confocal microscopy showed rapid phagocytic uptake of FITC-ZnONWs by macrophages. Exposure of macrophages or lung epithelial cells to ZnONWs induced the production of CCL2 and CCL11. Moreover, ZnONWs exposure induced both IL-6 and TNF-α production only in macrophages but not in LKR13 cells. Intratracheal instillation of ZnONWs in C57BL/6 mice induced a significant increase in the total numbers of immune cells in the broncho alveolar lavage fluid (BALFs) 2 days after instillation. Macrophages and eosinophils were the predominant cellular infiltrates of ZnONWs exposed mouse lungs. Similar cellular infiltrates were also observed in a mouse air-pouch model. Pro-inflammatory cytokines IL-6 and TNF-α as well as chemokines CCL11, and CCL2 were increased both in BALFs and air-pouch lavage fluids. These results suggest that exposure to ZnONWs may induce distinct inflammatory responses through phagocytic uptake and formation of soluble Zn2+ ions.
Collapse
Affiliation(s)
- Ruqaih S Alghsham
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Shuchismita R Satpathy
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Sobha R Bodduluri
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Bindu Hegde
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Venkatakrishna R Jala
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Waleed Twal
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Joseph A Burlison
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Mahendra Sunkara
- Department of Chemical Engineering, Conn Center for Renewable Energy, University of Louisville, Louisville, KY, United States
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
37
|
Wang G, Serkova NJ, Groman EV, Scheinman RI, Simberg D. Feraheme (Ferumoxytol) Is Recognized by Proinflammatory and Anti-inflammatory Macrophages via Scavenger Receptor Type AI/II. Mol Pharm 2019; 16:4274-4281. [PMID: 31556296 PMCID: PMC7513579 DOI: 10.1021/acs.molpharmaceut.9b00632] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Feraheme (ferumoxytol), a negatively charged, carboxymethyl dextran-coated ultrasmall superparamagnetic iron oxide nanoparticle (USPIO, 30 nm, -16 mV), is clinically approved as an iron supplement and is used off-label for magnetic resonance imaging (MRI) of macrophage-rich lesions, but the mechanism of recognition is not known. We investigated mechanisms of uptake of Feraheme by various types of macrophages in vitro and in vivo. The uptake by mouse peritoneal macrophages was not inhibited in complement-deficient serum. In contrast, the uptake of larger and less charged SPIO nanoworms (60 nm, -5 mV; 120 nm, -5 mV, respectively) was completely inhibited in complement deficient serum, which could be attributed to more C3 molecules bound per nanoparticle than Feraheme. The uptake of Feraheme in vitro was blocked by scavenger receptor (SR) inhibitor polyinosinic acid (PIA) and by antibody against scavenger receptor type A I/II (SR-AI/II). Antibodies against other SRs including MARCO, CD14, SR-BI, and CD11b had no effect on Feraheme uptake. Intraperitoneally administered PIA inhibited the peritoneal macrophage uptake of Feraheme in vivo. Nonmacrophage cells transfected with SR-AI plasmid efficiently internalized Feraheme but not noncharged ultrasmall SPIO of the same size (26 nm, -6 mV), suggesting that the anionic carboxymethyl groups of Feraheme are responsible for the SR-AI recognition. The uptake by nondifferentiated bone marrow derived macrophages (BMDM) and by BMDM differentiated into M1 (proinflammatory) and M2 (anti-inflammatory) types was efficiently inhibited by PIA and anti-SR-AI/II antibody. Interestingly, all BMDM types expressed similar levels of SR-AI/II. In conclusion, Feraheme is efficiently recognized via SR-AI/II but not via complement by different macrophage types. The recognition by the common phagocytic receptor has implications for specificity of imaging of macrophage subtypes.
Collapse
Affiliation(s)
- Guankui Wang
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie J. Serkova
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Departments of Radiology, Radiation Oncology, and Medicine/Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ernest V. Groman
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robert I. Scheinman
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Corresponding Author:
| |
Collapse
|
38
|
Llenas M, Sandoval S, Costa PM, Oró-Solé J, Lope-Piedrafita S, Ballesteros B, Al-Jamal KT, Tobias G. Microwave-Assisted Synthesis of SPION-Reduced Graphene Oxide Hybrids for Magnetic Resonance Imaging (MRI). NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1364. [PMID: 31554159 PMCID: PMC6835838 DOI: 10.3390/nano9101364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Magnetic resonance imaging (MRI) is a useful tool for disease diagnosis and treatment monitoring. Superparamagnetic iron oxide nanoparticles (SPION) show good performance as transverse relaxation (T2) contrast agents, thus facilitating the interpretation of the acquired images. Attachment of SPION onto nanocarriers prevents their agglomeration, improving the circulation time and efficiency. Graphene derivatives, such as graphene oxide (GO) and reduced graphene oxide (RGO), are appealing nanocarriers since they have both high surface area and functional moieties that make them ideal substrates for the attachment of nanoparticles. We have employed a fast, simple and environmentally friendly microwave-assisted approach for the synthesis of SPION-RGO hybrids. Different iron precursor/GO ratios were used leading to SPION, with a median diameter of 7.1 nm, homogeneously distributed along the RGO surface. Good relaxivity (r2*) values were obtained in MRI studies and no significant toxicity was detected within in vitro tests following GL261 glioma and J774 macrophage-like cells for 24 h with SPION-RGO, demonstrating the applicability of the hybrids as T2-weighted MRI contrast agents.
Collapse
Affiliation(s)
- Marina Llenas
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus de la UAB, 08193 Bellaterra (Barcelona), Spain.
| | - Stefania Sandoval
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus de la UAB, 08193 Bellaterra (Barcelona), Spain.
| | - Pedro M Costa
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK.
| | - Judith Oró-Solé
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus de la UAB, 08193 Bellaterra (Barcelona), Spain.
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Campus UAB, 08193 Bellaterra (Barcelona), Spain.
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Campus UAB, 08193 Bellaterra (Barcelona), Spain.
| | - Belén Ballesteros
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, Campus UAB, 08193 Bellaterra (Barcelona), Spain.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK.
| | - Gerard Tobias
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus de la UAB, 08193 Bellaterra (Barcelona), Spain.
| |
Collapse
|
39
|
Fathy MM, Fahmy HM, Balah AMM, Mohamed FF, Elshemey WM. Magnetic nanoparticles-loaded liposomes as a novel treatment agent for iron deficiency anemia: In vivo study. Life Sci 2019; 234:116787. [PMID: 31445028 DOI: 10.1016/j.lfs.2019.116787] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022]
Abstract
Iron deficiency anemia (IDA) is a major worldwide public health problem. This is due to its prevalence among infants, children, adolescents, pregnant and reproductive age women. Ferrous sulfate (FeSO4) is the first line therapy for iron IDA. Unfortunately, it is reported that FeSO4 suffers from low absorption rate in the body and itself exhibits severe side effects. Herein, iron oxide magnetic nanoparticles-loaded liposomes (LMNPs) are prepared, characterized and evaluated as a treatment regimen for IDA in Wistar rats (as an animal model). Iron oxide magnetic nanoparticles (MNPs) are prepared and loaded into liposomes using the thin film hydration method. The size of the prepared formulations is in the range 10-100 nm, thus it can avoid the reticular endothelial system (RES), and increased their blood circulation time. For in vivo assessment, thirty-five Wistar rats are divided into 5 groups (n = 7): negative control group, positive control group, and three groups treated with different iron formulations (FeSO4, MNPs and LMNPs). Anemia is induced in the anemic groups by the bleeding method and then treatment started with different iron compounds administrated orally for 13 days. Hematological parameters are followed up during the treatment period. Results indicate that, in the LMNPs group, the hematological parameters turn to normal values and the histopathological structures of the liver, spleen and kidney remain normal. This proves that liposome increases the bioavailability of MNPs. In conclusion, LMNPs demonstrate superiority as a therapeutic regimen for the treatment of IDA among the tested iron formulations.
Collapse
Affiliation(s)
- Mohamed M Fathy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | - Asmaa M M Balah
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Faten F Mohamed
- Pathology Department, Faculty of Veterinary Medicine, Giza 12211, Egypt
| | - Wael M Elshemey
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Department of Physics, Faculty of Science, Islamic University in Madinah, Saudi Arabia
| |
Collapse
|
40
|
Verçoza BR, Bernardo RR, Pentón-Madrigal A, Sinnecker JP, Rodrigues JC, S de Oliveira LA. Therapeutic potential of low-cost nanocarriers produced by green synthesis: macrophage uptake of superparamagnetic iron oxide nanoparticles. Nanomedicine (Lond) 2019; 14:2293-2313. [PMID: 31414612 DOI: 10.2217/nnm-2018-0500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: The primary goal of this work was to synthesize low-cost superparamagnetic iron oxide nanoparticles (SPIONs) with the aid of coconut water and evaluate the ability of macrophages to internalize them. Our motivation was to determine potential therapeutic applications in drug-delivery systems associated with magnetic hyperthermia. Materials & methods: We used the following characterization techniques: x-ray and electron diffractions, electron microscopy, spectrometry and magnetometry. Results: The synthesized SPIONs, roughly 4 nm in diameter, were internalized by macrophages, likely via endocytic/phagocytic pathways. They were randomly distributed throughout the cytoplasm and mainly located in membrane-bound compartments. Conclusion: Nanoparticles presented an elevated intrinsic loss power value and were not cytotoxic to mammalian cells. Thus, we suggest that low-cost SPIONs have great therapeutic potential.
Collapse
Affiliation(s)
- Brunno Rf Verçoza
- Núcleo Multidisciplinar de Pesquisas em Biologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-170, Brazil
| | - Robson R Bernardo
- Núcleo Multidisciplinar de Pesquisas em Biologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil.,Núcleo Multidisciplinar de Pesquisas em Nanotecnologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil
| | - Arbélio Pentón-Madrigal
- Facultad de Física, IMRE, Universidad de La Habana, San Lazaro y L, C. Habana, CP 10400, Cuba
| | - João P Sinnecker
- Centro Brasileiro de Pesquisas Físicas, Rua Xavier Sigaud 150, Rio de Janeiro, RJ, 22290-180, Brazil
| | - Juliany Cf Rodrigues
- Núcleo Multidisciplinar de Pesquisas em Biologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, 21941-170, Brazil
| | - Luiz Augusto S de Oliveira
- Núcleo Multidisciplinar de Pesquisas em Biologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil.,Núcleo Multidisciplinar de Pesquisas em Nanotecnologia, Campus Prof. Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rodovia Washington Luiz, km 105. 25240-005, Duque de Caxias, RJ, Brazil
| |
Collapse
|
41
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
42
|
Rabel M, Warncke P, Grüttner C, Bergemann C, Kurland HD, Müller R, Dugandžić V, Thamm J, Müller FA, Popp J, Cialla-May D, Fischer D. Simulation of the long-term fate of superparamagnetic iron oxide-based nanoparticles using simulated biological fluids. Nanomedicine (Lond) 2019; 14:1681-1706. [DOI: 10.2217/nnm-2018-0382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To simulate the stability and degradation of superparamagnetic iron oxide nanoparticles (MNP) in vitro as part of their life cycle using complex simulated biological fluids. Materials & methods: A set of 13 MNP with different polymeric or inorganic shell materials was synthesized and characterized regarding stability and degradation of core and shell in simulated biological fluids. Results: All MNP formulations showed excellent stability during storage and in simulated body fluid. In endosomal/lysosomal media the degradation behavior depended on shell characteristics (e.g., charge, acid-base character) and temperature enabling the development of an accelerated stress test protocol. Conclusion: Kinetics of transformations depending on the MNP type could be established to define structure-activity relationships as prediction model for rational particle design.
Collapse
Affiliation(s)
- Martin Rabel
- Pharmaceutical Technology & Biopharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Paul Warncke
- Pharmaceutical Technology & Biopharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | | | - Heinz-Dieter Kurland
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Robert Müller
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Vera Dugandžić
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Jana Thamm
- Pharmaceutical Technology & Biopharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743 Jena, Germany
| | - Frank A. Müller
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Dana Cialla-May
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Dagmar Fischer
- Pharmaceutical Technology & Biopharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743 Jena, Germany
| |
Collapse
|
43
|
Jiang Z, Lin C, Liu H, Feng J, Zheng Z, Cai S. Specific biological responses following dextran-coated ultra-small superparamagnetic particles of iron oxides administration. Nanomedicine (Lond) 2019; 14:1371-1386. [PMID: 31215328 DOI: 10.2217/nnm-2018-0433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The potential bio-related risks of dextran-coated ultra-small superparamagnetic particles of iron oxides (D-USPIO) were assessed. Materials & methods: Metabolic responses of D-USPIO in BALB/C mice were obtained using 1H-NMR-based metabolomic strategy combined with the traditional biochemical assay. Results: The metabolomic analyses of biological fluids (plasma and urine) and organs (liver, kidney and spleen) indicated that the disturbance, impairment and recovery of the physiological functions were related to the metabolic response to D-USPIO. The correlations between the biofluids and tissue metabolomes described the specific metabolic information of D-USPIO on their in vivo transportation, absorption, biodistribution and excretion. Conclusion: Metabolomic analysis provides preliminary validation for the use of D-USPIO in clinical medicine, and the results help to understand the potential adverse effects of the similar bio-nanomaterials further serve to their synthesis optimization and biocompatibility improvement.
Collapse
Affiliation(s)
- Zhiming Jiang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma & Magnetic Resonance, Xiamen University, Xiamen 361005, PR China
| | - Chenghong Lin
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma & Magnetic Resonance, Xiamen University, Xiamen 361005, PR China
| | - Huili Liu
- State Key Laboratory of Magnetic Resonance & Atomic & Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics & Mathematics, Chinese Academy of Sciences, Wuhan 430071, PR China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma & Magnetic Resonance, Xiamen University, Xiamen 361005, PR China
| | - Zhenyao Zheng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma & Magnetic Resonance, Xiamen University, Xiamen 361005, PR China
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma & Magnetic Resonance, Xiamen University, Xiamen 361005, PR China
| |
Collapse
|
44
|
Trujillo-Alonso V, Pratt EC, Zong H, Lara-Martinez A, Kaittanis C, Rabie MO, Longo V, Becker MW, Roboz GJ, Grimm J, Guzman ML. FDA-approved ferumoxytol displays anti-leukaemia efficacy against cells with low ferroportin levels. NATURE NANOTECHNOLOGY 2019; 14:616-622. [PMID: 30911166 PMCID: PMC6554053 DOI: 10.1038/s41565-019-0406-1] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 02/14/2019] [Indexed: 05/17/2023]
Abstract
Acute myeloid leukaemia is a fatal disease for most patients. We have found that ferumoxytol (Feraheme), an FDA-approved iron oxide nanoparticle for iron deficiency treatment, demonstrates an anti-leukaemia effect in vitro and in vivo. Using leukaemia cell lines and primary acute myeloid leukaemia patient samples, we show that low expression of the iron exporter ferroportin results in a susceptibility of these cells via an increase in intracellular iron from ferumoxytol. The reactive oxygen species produced by free ferrous iron lead to increased oxidative stress and cell death. Ferumoxytol treatment results in a significant reduction of disease burden in a murine leukaemia model and patient-derived xenotransplants bearing leukaemia cells with low ferroportin expression. Our findings show how a clinical nanoparticle previously considered largely biologically inert could be rapidly incorporated into clinical trials for patients with leukaemia with low ferroportin levels.
Collapse
Affiliation(s)
- Vicenta Trujillo-Alonso
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Edwin C Pratt
- Department of Pharmacology, Weill Cornell Graduate School, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hongliang Zong
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Andres Lara-Martinez
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Charalambos Kaittanis
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohamed O Rabie
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Valerie Longo
- Small-Animal Imaging Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael W Becker
- Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Gail J Roboz
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jan Grimm
- Department of Pharmacology, Weill Cornell Graduate School, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Monica L Guzman
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Graduate School, New York, NY, USA.
| |
Collapse
|
45
|
Wei Q, Wang J, Shi W, Zhang B, Jiang H, Du M, Mei H, Hu Y. Improved in vivo detection of atherosclerotic plaques with a tissue factor-targeting magnetic nanoprobe. Acta Biomater 2019; 90:324-336. [PMID: 30954623 DOI: 10.1016/j.actbio.2019.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 11/19/2022]
Abstract
Rupture of atherosclerotic plaques causes acute cardiovascular and cerebrovascular pathology. Tissue factor (TF) is a key factor that affects the development of atherosclerotic plaques and the formation of thrombus and thus constitutes a potential target for the detection of atherosclerotic plaques. In this study, the conjugation of the fusion protein 'enhanced green fluorescent protein with the first epidermal growth factor domain' (EGFP-EGF1) and superparamagnetic iron oxide nanoparticles (EGFP-EGF1-SPIONs) was explored for molecular imaging of TF-positive atherosclerotic plaques. EGFP-EGF1-SPIONs showed improved accuracy, superior contrast effects, and better cytocompatibility compared with common contrast agents in the detection of atherosclerotic plaques of apolipoprotein E knockout (ApoE-/-) mice using magnetic resonance imaging. In conclusion, EGFP-EGF1-SPION is a promising TF-targeting nanoprobe to precisely and specifically detect atherosclerotic plaques, which may improve molecular imaging diagnosis of cardiovascular and cerebrovascular events for the comprehensive evaluation of atherosclerosis. STATEMENT OF SIGNIFICANCE: Traditional methods can only display the status of atherosclerosis, but not forecast the progress of lesions efficiently. It remains challenging to evaluate the plaques specifically and sensitively. In this study, we constructed a tissue factor-targeted magnetic nanoprobe to specifically detect plaques by magnetic resonance imaging in vivo, which will improve the diagnostic technology for atherosclerotic plaques and offer molecular level guidance to treat atherosclerosis. Furthermore, this strategy has critical clinical significance on prevention, diagnosis and therapeutic evaluation of cardio-cerebral vascular events.
Collapse
Affiliation(s)
- Qiuzhe Wei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China
| | - Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
46
|
Ovais M, Guo M, Chen C. Tailoring Nanomaterials for Targeting Tumor-Associated Macrophages. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1808303. [PMID: 30883982 DOI: 10.1002/adma.201808303] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/07/2019] [Indexed: 05/17/2023]
Abstract
Advances in the field of nanotechnology together with an increase understanding of tumor immunology have paved the way for the development of more personalized cancer immuno-nanomedicines. Nanovehicles, due to their specific physicochemical properties, are emerging as key translational moieties in tackling tumor-promoting, M2-like tumor-associated macrophages (TAMs). Cancer immuno-nanomedicines target TAMs primarily by blocking M2-like TAM survival or affecting their signaling cascades, restricting macrophage recruitment to tumors and re-educating tumor-promoting M2-like TAMs to the tumoricidal, M1-like phenotype. Here, the TAM effector mechanisms and strategies for targeting TAMs are summarized, followed by a focus on the mechanistic considerations in the development of novel immuno-nanomedicines. Furthermore, imaging TAMs with nanoparticles so as to forecast a patient's clinical outcome, describing treatment options, and observing therapy responses is also discussed. At present, strategies that target TAMs are being investigated not only at the basic research level but also in early clinical trials. The significance of TAM-targeting biomaterials is highlighted, with the goal of facilitating future clinical translation.
Collapse
Affiliation(s)
- Muhammad Ovais
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
47
|
Janko C, Ratschker T, Nguyen K, Zschiesche L, Tietze R, Lyer S, Alexiou C. Functionalized Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as Platform for the Targeted Multimodal Tumor Therapy. Front Oncol 2019; 9:59. [PMID: 30815389 PMCID: PMC6382019 DOI: 10.3389/fonc.2019.00059] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/21/2019] [Indexed: 01/21/2023] Open
Abstract
Standard cancer treatments involve surgery, radiotherapy, chemotherapy, and immunotherapy. In clinical practice, the respective drugs are applied orally or intravenously leading to their systemic circulation in the whole organism. For chemotherapeutics or immune modulatory agents, severe side effects such as immune depression or autoimmunity can occur. At the same time the intratumoral drug doses are often too low for effective cancer therapy. Since monotherapies frequently cannot cure cancer, due to their synergistic effects multimodal therapy concepts are applied to enhance treatment efficacy. The targeted delivery of drugs to the tumor by employment of functionalized nanoparticles might be a promising solution to overcome these challenges. For multimodal therapy concepts and individualized patient care nanoparticle platforms can be functionalized with compounds from various therapeutic classes (e.g. radiosensitizers, phototoxic drugs, chemotherapeutics, immune modulators). Superparamagnetic iron oxide nanoparticles (SPIONs) as drug transporters can add further functionalities, such as guidance or heating by external magnetic fields (Magnetic Drug Targeting or Magnetic Hyperthermia), and imaging-controlled therapy (Magnetic Resonance Imaging).
Collapse
Affiliation(s)
- Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Teresa Ratschker
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany.,Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Khanh Nguyen
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany.,Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa Zschiesche
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany.,Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
48
|
Radha G, Venkatesan B, Rajashree P, Vellaichamy E, Balakumar S. Insights into the apatite mineralization potential of thermally processed nanocrystalline Ca10−xFex(PO4)6(OH)2. NEW J CHEM 2019. [DOI: 10.1039/c8nj03579b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The thermal treatment of Ca10−xFex(PO4)6(OH)2 at different temperatures had an effect on the mineralization potential under non-cellular and cellular conditions by releasing its bioactive ions at optimal or excessive levels.
Collapse
Affiliation(s)
- G. Radha
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600025
- India
| | | | - P. Rajashree
- Centre for Advanced Study in Crystallography and Biophysics
- University of Madras
- Chennai – 600025
- India
| | | | - S. Balakumar
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600025
- India
| |
Collapse
|
49
|
Casset A, Jouhannaud J, Garofalo A, Spiegelhalter C, Nguyen DV, Felder-Flesch D, Pourroy G, Pons F. Macrophage functionality and homeostasis in response to oligoethyleneglycol-coated IONPs: Impact of a dendritic architecture. Int J Pharm 2018; 556:287-300. [PMID: 30557682 DOI: 10.1016/j.ijpharm.2018.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022]
Abstract
The engineering of iron oxide nanoparticles (IONPs) for biomedical use has received great interest over the past decade. In the present study we investigated the biocompatibility of IONPs grafted with linear (2P) or generation 1 (2PG1) or 2 (2PG2) dendronized oligoethyleneglycol units in THP-1-derived macrophages. To evaluate IONP effects on cell functionality and homeostasis, mitochondrial function (MTT assay), membrane permeability (LDH release), inflammation (IL-8), oxidative stress (reduced glutathione, GSH), NLRP3 inflammasome activation (IL-1β) and nanoparticle cellular uptake (intracellular iron content) were quantified after a 4-h or 24-h cell exposure to increasing IONP concentrations (0-300 µg Fe/mL). IONPs coated with a linear molecule, NP10COP@2P, were highly taken up by cells and induced significant dose-dependent IL-8 release, oxidative stress and NLRP3 inflammasome activation. In comparison, IONPs coated with dendrons of generation 1 (NP10COP@2PG1) and 2 (NP10COP@2PG2) exhibited better biocompatibility. Effect of the dendritic architecture of the surface coating was investigated in a kinetic experiment involving cell short-term exposure (30 min or 1 h 30) to the two dendronized IONPs. NP10COP@2PG2 disrupted cellular homeostasis (LDH release, IL-1β and IL-8 secretion) to a greater extend than NP10COP@2PG1, which makes this last IONP the best candidate as MRI contrast or theranostic agent.
Collapse
Affiliation(s)
- Anne Casset
- Université de Strasbourg, CNRS, CAMB UMR 7199, F-67000 Strasbourg, France.
| | - Julien Jouhannaud
- Université de Strasbourg, CNRS, Institut de Physique et Chimie des Matériaux Strasbourg, UMR 7504, F-67000 Strasbourg, France
| | - Antonio Garofalo
- Université de Strasbourg, CNRS, Institut de Physique et Chimie des Matériaux Strasbourg, UMR 7504, F-67000 Strasbourg, France
| | - Coralie Spiegelhalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, CNRS, Université de Strasbourg, F-67404 Illkirch, France
| | - Dinh-Vu Nguyen
- Université de Strasbourg, CNRS, Institut de Physique et Chimie des Matériaux Strasbourg, UMR 7504, F-67000 Strasbourg, France
| | - Delphine Felder-Flesch
- Université de Strasbourg, CNRS, Institut de Physique et Chimie des Matériaux Strasbourg, UMR 7504, F-67000 Strasbourg, France
| | - Geneviève Pourroy
- Université de Strasbourg, CNRS, Institut de Physique et Chimie des Matériaux Strasbourg, UMR 7504, F-67000 Strasbourg, France
| | - Françoise Pons
- Université de Strasbourg, CNRS, CAMB UMR 7199, F-67000 Strasbourg, France
| |
Collapse
|
50
|
Abstract
The recent clinical success of cancer immunotherapy has renewed interest in the development of tools to image the immune system. In general, immunotherapies attempt to enable the body's own immune cells to seek out and destroy malignant disease. Molecular imaging of the cells and molecules that regulate immunity could provide unique insight into the mechanisms of action, and failure, of immunotherapies. In this article, we will provide a comprehensive overview of the current state-of-the-art immunoimaging toolbox with a focus on imaging strategies and their applications toward immunotherapy.
Collapse
Affiliation(s)
- Aaron T Mayer
- Department of Bioengineering, Stanford University, Stanford, California; and
| | - Sanjiv S Gambhir
- Department of Bioengineering, Stanford University, Stanford, California; and
- Department of Radiology, Department of Materials Science and Engineering, Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| |
Collapse
|