1
|
Sharma P, Hoorn D, Aitha A, Breier D, Peer D. The immunostimulatory nature of mRNA lipid nanoparticles. Adv Drug Deliv Rev 2024; 205:115175. [PMID: 38218350 DOI: 10.1016/j.addr.2023.115175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024]
Abstract
mRNA-Lipid nanoparticles (LNPs) are at the forefront of global medical research. With the development of mRNA-LNP vaccines to combat the COVID-19 pandemic, the clinical potential of this platform was unleashed. Upon administering 16 billion doses that protected billions of people, it became clear that a fraction of them witnessed mild and in some cases even severe adverse effects. Therefore, it is paramount to define the safety along with the therapeutic efficacy of the mRNA-LNP platform for the successful translation of new genetic medicines based on this technology. While mRNA was the effector molecule of this platform, the ionizable lipid component of the LNPs played an indispensable role in its success. However, both of these components possess the ability to induce undesired immunostimulation, which is an area that needs to be addressed systematically. The immune cell agitation caused by this platform is a two-edged sword as it may prove beneficial for vaccination but detrimental to other applications. Therefore, a key challenge in advancing the mRNA-LNP drug delivery platform from bench to bedside is understanding the immunostimulatory behavior of these components. Herein, we provide a detailed overview of the structural modifications and immunogenicity of synthetic mRNA. We discuss the effect of ionizable lipid structure on LNP functionality and offer a mechanistic overview of the ability of LNPs to elicit an immune response. Finally, we shed some light on the current status of this technology in clinical trials and discuss a few challenges to be addressed to advance the field.
Collapse
Affiliation(s)
- Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Daniek Hoorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Anjaiah Aitha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
2
|
Padilla S, Prado R, Anitua E. An evolutionary history of F12 gene: Emergence, loss, and vulnerability with the environment as a driver. Bioessays 2023; 45:e2300077. [PMID: 37750435 DOI: 10.1002/bies.202300077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
In the context of macroevolutionary transitions, environmental changes prompted vertebrates already bearing genetic variations to undergo gradual adaptations resulting in profound anatomical, physiological, and behavioral adaptations. The emergence of new genes led to the genetic variation essential in metazoan evolution, just as was gene loss, both sources of genetic variation resulting in adaptive phenotypic diversity. In this context, F12-coding protein with defense and hemostatic roles emerged some 425 Mya, and it might have contributed in aquatic vertebrates to the transition from water-to-land. Conversely, the F12 loss in marine, air-breathing mammals like cetaceans has been associated with phenotypic adaptations in some terrestrial mammals in their transition to aquatic lifestyle. More recently, the advent of technological innovations in western lifestyle with blood-contacting devices and harmful environmental nanoparticles, has unfolded new roles of FXII. Environment operates as either a positive or a relaxed selective pressure on genes, and consequently genes are selected or lost. FXII, an old dog facing environmental novelties can learn new tricks and teach us new therapeutic avenues.
Collapse
Affiliation(s)
- Sabino Padilla
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Prado
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Eduardo Anitua
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
3
|
Giambelluca M, Markova E, Louet C, Steinkjer B, Sundset R, Škalko-Basnet N, Hak S. Liposomes - Human phagocytes interplay in whole blood: effect of liposome design. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 54:102712. [PMID: 37838100 DOI: 10.1016/j.nano.2023.102712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/16/2023]
Abstract
Nanomedicine holds immense potential for therapeutic manipulation of phagocytic immune cells. However, in vitro studies often fail to accurately translate to the complex in vivo environment. To address this gap, we employed an ex vivo human whole-blood assay to evaluate liposome interactions with immune cells. We systematically varied liposome size, PEG-surface densities and sphingomyelin and ganglioside content. We observed differential uptake patterns of the assessed liposomes by neutrophils and monocytes, emphasizing the importance of liposome design. Interestingly, our results aligned closely with published in vivo observations in mice and patients. Moreover, liposome exposure induced changes in cytokine release and cellular responses, highlighting the potential modulation of immune system. Our study highlights the utility of human whole-blood models in assessing nanoparticle-immune cell interactions and provides insights into liposome design for modulating immune responses.
Collapse
Affiliation(s)
- Miriam Giambelluca
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Elena Markova
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Claire Louet
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Bjørg Steinkjer
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rune Sundset
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Sjoerd Hak
- Department of Biotechnology and Nanomedicine, SINTEF Industry and Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
4
|
Christodoulides A, Hall A, Alves NJ. Exploring microplastic impact on whole blood clotting dynamics utilizing thromboelastography. Front Public Health 2023; 11:1215817. [PMID: 37521965 PMCID: PMC10372794 DOI: 10.3389/fpubh.2023.1215817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
This study investigates the influence of microplastics on blood clotting. It addresses the lack of comprehensive research on the effects of microplastic size and surface modification on clotting dynamics in human whole blood. Thromboelastography was used to examine aminated (aPS), carboxylated (cPS), and non-functionalized (nPS) polystyrene particles with sizes of 50, 100, and 500 nm. Results show that cPS consistently activated the clotting cascade, demonstrating increased fibrin polymerization rates, and enhanced clot strength in a size and concentration-dependent manner. nPS had minimal effects on clotting dynamics except for 50 nm particles at the lowest concentration. The clotting effects of aPS (100 nm particles) resembled those of cPS but were diminished in the 500 nm aPS group. These findings emphasize the importance of microplastic surface modification, size, concentration, and surface area on in-vitro whole blood clotting dynamics.
Collapse
Affiliation(s)
- Alexei Christodoulides
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Abigail Hall
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nathan J. Alves
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
5
|
Gerogianni A, Bal M, Mohlin C, Woodruff TM, Lambris JD, Mollnes TE, Sjöström DJ, Nilsson PH. In vitro evaluation of iron oxide nanoparticle-induced thromboinflammatory response using a combined human whole blood and endothelial cell model. Front Immunol 2023; 14:1101387. [PMID: 37081885 PMCID: PMC10111002 DOI: 10.3389/fimmu.2023.1101387] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) are widely used in diagnostic and therapeutic settings. Upon systemic administration, however, they are rapidly recognized by components of innate immunity, which limit their therapeutic capacity and can potentially lead to adverse side effects. IONPs were previously found to induce the inflammatory response in human whole blood, including activation of the complement system and increased secretion of cytokines. Here, we investigated the thromboinflammatory response of 10-30 nm IONPs in lepirudin anticoagulated whole blood in interplay with endothelial cells and evaluated the therapeutic effect of applying complement inhibitors to limit adverse effects related to thromboinflammation. We found that IONPs induced complement activation, primarily at the C3-level, in whole blood incubated for up to four hours at 37°C with and without human microvascular endothelial cells. Furthermore, IONPs mediated a strong thromboinflammatory response, as seen by the significantly increased release of 21 of the 27 analyzed cytokines (p<0.05). IONPs also significantly increased cell-activation markers of endothelial cells [ICAM-1 (p<0.0001), P/E-selectin (p<0.05)], monocytes, and granulocytes [CD11b (p<0.001)], and platelets [CD62P (p<0.05), CD63 (p<0.05), NAP-2 (p<0.01), PF4 (p<0.05)], and showed cytotoxic effects, as seen by increased LDH (p<0.001) and heme (p<0.0001) levels. We found that inflammation and endothelial cell activation were partly complement-dependent and inhibition of complement at the level of C3 by compstatin Cp40 significantly attenuated expression of ICAM-1 (p<0.01) and selectins (p<0.05). We show that complement activation plays an important role in the IONPs-induced thromboinflammatory response and that complement inhibition is promising in improving IONPs biocompatibility.
Collapse
Affiliation(s)
- Alexandra Gerogianni
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
| | - Melissa Bal
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
| | - Camilla Mohlin
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
| | - Trent M. Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Research Laboratory, Nordland Hospital, Bodo, Norway
| | - Dick J. Sjöström
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
| | - Per H. Nilsson
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- *Correspondence: Per H. Nilsson,
| |
Collapse
|
6
|
Fromell K, Johansson U, Abadgar S, Bourzeix P, Lundholm L, Elihn K. The effect of airborne Palladium nanoparticles on human lung cells, endothelium and blood - A combinatory approach using three in vitro models. Toxicol In Vitro 2023; 89:105586. [PMID: 36931534 DOI: 10.1016/j.tiv.2023.105586] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/24/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
A better understanding of the mechanisms behind adverse health effects caused by airborne fine particles and nanoparticles (NP) is essential to improve risk assessment and identification the most critical particle exposures. While the use of automobile catalytic converters is decreasing the exhausts of harmful gases, concentrations of fine airborne particles and nanoparticles (NPs) from catalytic metals such as Palladium (Pd) are reaching their upper safe level. Here we used a combinatory approach with three in vitro model systems to study the toxicity of Pd particles, to infer their potential effects on human health upon inhalation. The three model systems are 1) a lung system with human lung cells (ALI), 2) an endothelial cell system and 3) a human whole blood loop system. All three model systems were exposed to the exact same type of Pd NPs. The ALI lung cell exposure system showed a clear reduction in cell growth from 24 h onwards and the effect persisted over a longer period of time. In the endothelial cell model, Pd NPs induced apoptosis, but not to the same extent as the most aggressive types of NPs such as TiO2. Similarly, Pd triggered clear coagulation and contact system activation but not as forcefully as the highly thrombogenic TiO2 NPs. In summary, we show that our 3-step in vitro model of the human lung and surrounding vessels can be a useful tool for studying pathological events triggered by airborne fine particles and NPs.
Collapse
Affiliation(s)
- Karin Fromell
- Department of Immunology, Genetics and Pathology, Rudbeck laboratory C5:3, Uppsala university, SE-751 85 Uppsala, Sweden.
| | - Ulrika Johansson
- Department of Immunology, Genetics and Pathology, Rudbeck laboratory C5:3, Uppsala university, SE-751 85 Uppsala, Sweden; Linnæus Centre for Biomaterials Chemistry, Linnæus University, SE-391 82 Kalmar, Sweden
| | - Sophia Abadgar
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden; Department of Environmental Science, Stockholm University, 106 91 Stockholm, Sweden
| | - Pauline Bourzeix
- Department of Immunology, Genetics and Pathology, Rudbeck laboratory C5:3, Uppsala university, SE-751 85 Uppsala, Sweden
| | - Lovisa Lundholm
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Karine Elihn
- Department of Environmental Science, Stockholm University, 106 91 Stockholm, Sweden
| |
Collapse
|
7
|
Suzuki H, Adler A, Huang T, Kuramochi A, Ohba Y, Sato Y, Nakamura N, Manivel VA, Ekdahl KN, Nilsson B, Ishihara K, Teramura Y. Impact of spontaneous liposome modification with phospholipid polymer-lipid conjugates on protein interactions. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:845-857. [PMID: 36518982 PMCID: PMC9744213 DOI: 10.1080/14686996.2022.2146466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
Liposome surface coating has been studied to avoid the immunological responses caused by the complement system, and alternative materials to poly(ethylene glycol) (PEG) have been explored recently since the production of anti-PEG IgM antibodies has been found in humans. We previously reported a liposome coating with poly(2-methacryloyloxyethyl phosphorylcholine) (poly(MPC))-conjugated lipids (PMPC-lipids) and demonstrated its protective effect on blood protein interactions. Here, we attempted to modify the liposome surface by exogenously adding PMPC-lipids, which were spontaneously incorporated into the outer membrane via hydrophobic interactions. The polymerization degree of the PMPC segment was regulated from 10 to 100. The incorporated ratio of PMPC-lipid increased with a decrease in the degree of PMPC polymerization. Due to surface modification with PMPC-lipids, increase in the length of the PMPC-chain increased the size of the liposomes. The modified liposomes were kept stable for 14 d in terms of their size, polydispersity, and surface properties, where approximately 70% of PMPC-lipids were incorporated into the liposome surface. We demonstrated that liposome surface modification with PMPC-lipids can inhibit protein adsorption when exposed to serum, regardless of the degree of polymerization of PMPC. In addition, the PMPC-lipid modified surface was not recognized by the anti-PEG IgM antibody, whereas PEG-lipid was recognized by the antibody. Thus, we successfully fabricated an inert liposome surface via spontaneous modification with PMPC-lipids, where only the outer bilayer surface was modified. This technique can be available for full loading of water-soluble active pharmaceutical ingredient inside the modified liposome.
Collapse
Affiliation(s)
- Haruna Suzuki
- Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Anna Adler
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Tianwei Huang
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Akiko Kuramochi
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Yoshiro Ohba
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Yuya Sato
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Naoko Nakamura
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Vivek Anand Manivel
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
| | - Kazuhiko Ishihara
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Yuji Teramura
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
- Master’s/Doctoral Program in Life Science Innovation (T-LSI), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
8
|
Lira AL, Mina N, Bonturi CR, Nogueira RS, Torquato RJS, Oliva MLV, Sousa AA. Anionic Ultrasmall Gold Nanoparticles Bind to Coagulation Factors and Disturb Normal Hemostatic Balance. Chem Res Toxicol 2022; 35:1558-1569. [PMID: 36018252 DOI: 10.1021/acs.chemrestox.2c00190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ultrasmall gold nanoparticles (usNPs) and nanoclusters are an emerging class of nanomaterials exhibiting distinctive physicochemical properties and in vivo behaviors. Although understanding the interactions of usNPs with blood components is of fundamental importance to advance their clinical translation, currently, little is known about the way that usNPs interact with the hemostatic system. This study describes the effects of a model anionic p-mercaptobenzoic acid-coated usNP on the coagulation cascade, with particular emphasis on the contact pathway. It is found that in a purified system, the anionic usNPs bind to and activate factor XII (FXII). The formed usNP-FXII complexes are short-lived (residence time of ∼10 s) and characterized by an affinity constant of ∼200 nM. In human plasma, the anionic usNPs activate the contact pathway and promote coagulation. The usNPs also exhibit anticoagulant activity in plasma by interfering with the thrombin-mediated cleavage of fibrinogen. Taken together, these findings establish that anionic usNPs can disturb the normal hemostatic balance, which in turn may hinder their clinical translation. Finally, it is shown that usNPs can be designed to be nearly inert in plasma by surface coating with the natural peptide glutathione.
Collapse
Affiliation(s)
- André L Lira
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Natasha Mina
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Camila R Bonturi
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Ruben S Nogueira
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Ricardo J S Torquato
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Maria Luiza V Oliva
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Alioscka A Sousa
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| |
Collapse
|
9
|
Evolutionary Insight into Immunothrombosis as a Healing Mechanism. Int J Mol Sci 2022; 23:ijms23158346. [PMID: 35955499 PMCID: PMC9368803 DOI: 10.3390/ijms23158346] [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: 06/20/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Both invertebrates and vertebrates possess a cluster of immediate and local wound-sealing, pathogen-killing, and tissue healing responses known as immunoclotting and immunothrombosis, respectively, to cope with two life-threatening emergencies, namely, bleeding and microbial invasion. Despite their convergence in function, immunoclotting and immunothrombosis are deployed by different blood cells and intravascular multidomain proteins. In vertebrates, these proteins share some domains with intrinsic chemical affinities useful in generating cooperative networks such as pathogen and damage pattern recognition molecules. Moreover, many of the proteins involved in coagulation and fibrinolysis in humans are multifunctional molecules playing roles in other processes from inflammation to healing and beyond. In our modern society, however, the interaction of activated intravascular allosteric proteins with one another and with blood cells entails vulnerabilities posing a biological paradox: intravascular proteins that locally operate as tissue repair enhancers can nevertheless generate pathogenic processes by acting systemically. In this manuscript, we contextualize and frame the coagulation system and hemostasis through an evolutionary time scale, illustrating their role as dual players in the defense against exsanguination and pathogens while significantly influencing wound healing.
Collapse
|
10
|
Tran HDN, Moonshi SS, Xu ZP, Ta HT. Influence of nanoparticles on the haemostatic balance: between thrombosis and haemorrhage. Biomater Sci 2021; 10:10-50. [PMID: 34775503 DOI: 10.1039/d1bm01351c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Maintenance of a delicate haemostatic balance or a balance between clotting and bleeding is critical to human health. Irrespective of administration route, nanoparticles can reach the bloodstream and might interrupt the haemostatic balance by interfering with one or more components of the coagulation, anticoagulation, and fibrinolytic systems, which potentially lead to thrombosis or haemorrhage. However, inadequate understanding of their effects on the haemostatic balance, along with the fact that most studies mainly focus on the functionality of nanoparticles while forgetting or leaving behind their risk to the body's haemostatic balance, is a major concern. Hence, our review aims to provide a comprehensive depiction of nanoparticle-haemostatic balance interactions, which has not yet been covered. The synergistic roles of cells and plasma factors participating in haemostatic balance are presented. Possible interactions and interference of each type of nanoparticle with the haemostatic balance are comprehensively discussed, particularly focusing on the underlying mechanisms. Interactions of nanoparticles with innate immunity potentially linked to haemostasis are mentioned. Various physicochemical characteristics that influence the nanoparticle-haemostatic balance are detailed. Challenges and future directions are also proposed. This insight would be valuable for the establishment of nanoparticles that can either avoid unintended interference with the haemostatic balance or purposely downregulate/upregulate its key components in a controlled manner.
Collapse
Affiliation(s)
- Huong D N Tran
- Queensland Micro- and Nanotechnology, Griffith University, Nathan, Queensland 4111, Australia. .,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | | | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hang Thu Ta
- Queensland Micro- and Nanotechnology, Griffith University, Nathan, Queensland 4111, Australia. .,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
11
|
Padilla S, Nurden AT, Prado R, Nurden P, Anitua E. Healing through the lens of immunothrombosis: Biology-inspired, evolution-tailored, and human-engineered biomimetic therapies. Biomaterials 2021; 279:121205. [PMID: 34710794 DOI: 10.1016/j.biomaterials.2021.121205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
Evolution, from invertebrates to mammals, has yielded and shaped immunoclotting as a defense and repair response against trauma and infection. This mosaic of immediate and local wound-sealing and pathogen-killing mechanisms results in survival, restoration of homeostasis, and tissue repair. In mammals, immunoclotting has been complemented with the neuroendocrine system, platelets, and contact system among other embellishments, adding layers of complexity through interconnecting blood-born proteolytic cascades, blood cells, and the neuroendocrine system. In doing so, immunothrombosis endows humans with survival advantages, but entails vulnerabilities in the current unprecedented and increasingly challenging environment. Immunothrombosis and tissue repair appear to go hand in hand with common mechanisms mediating both processes, a fact that is underlined by recent advances that are deciphering the mechanisms of the repair process and of the biochemical pathways that underpins coagulation, hemostasis and thrombosis. This review is intended to frame both the universal aspects of tissue repair and the therapeutic use of autologous fibrin matrix as a biology-as-a-drug approach in the context of the evolutionary changes in coagulation and hemostasis. In addition, we will try to shed some light on the molecular mechanisms underlying the use of the autologous fibrin matrix as a biology-inspired, evolution-tailored, and human-engineered biomimetic therapy.
Collapse
Affiliation(s)
- Sabino Padilla
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| | - Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| | - Roberto Prado
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Paquita Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| | - Eduardo Anitua
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| |
Collapse
|
12
|
Maisha N, Naik N, Okesola M, Coombs T, Zilberberg R, Pandala N, Lavik E. Engineering PEGylated Polyester Nanoparticles to Reduce Complement-Mediated Infusion Reaction. Bioconjug Chem 2021; 32:2154-2166. [PMID: 34499487 DOI: 10.1021/acs.bioconjchem.1c00339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Translation of intravenously administered nanomaterials to the clinic is limited due to adverse infusion reactions. While these reactions are infrequent, with up to 10% prone to experiencing infusion reactions, the reactions can be severe and life-threatening. One of the innate immune pathways, the complement activation pathway, plays a significant role in mediating this response. Nanoparticle surface properties are a relevant design feature, as they control the blood proteins the nanoparticles interact with and allow the nanoparticles to evade the immune reaction. PEGylation of nanosurfaces is critical in improving the blood circulation of nanoparticles and reducing opsonization. Our goal was to understand whether modifying the surface architecture by varying the PEG density and architecture can impact the complement response in vitro. We utilized block copolymers of poly(lactic acid)-b-poly(ethylene glycol) prepared with poly(ethylene glycol) macroinitiators of molecular weights 3400 and 5000 Da. Tracking the complement biomarker C5a, we monitored the impact of changing PEGylation of the nanoparticles. We also investigated how the changing PEG length on the nanoparticle surface impacts further strengthening the stealth properties. Lastly, we determined which cytokines change upon blood incubation with nanoparticles in vitro to understand the extent to which inflammation may occur and the crosstalk between the complement and immune responses. Increasing PEGylation reduced the generation of complement-mediated anaphylatoxin C5a in vitro, with 5000 Da PEG more effectively reducing levels of C5a generated compared to 3400 Da PEG. The insights gathered regarding the impact of PEG density and PEG chain length would be critical in developing stealth nanoparticles that do not lead to infusion reactions upon intravenous administration.
Collapse
Affiliation(s)
- Nuzhat Maisha
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Nidhi Naik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Mawuyon Okesola
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Tobias Coombs
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Rose Zilberberg
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Narendra Pandala
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Erin Lavik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| |
Collapse
|
13
|
Mannes M, Schmidt CQ, Nilsson B, Ekdahl KN, Huber-Lang M. Complement as driver of systemic inflammation and organ failure in trauma, burn, and sepsis. Semin Immunopathol 2021; 43:773-788. [PMID: 34191093 PMCID: PMC8243057 DOI: 10.1007/s00281-021-00872-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023]
Abstract
Complement is one of the most ancient defense systems. It gets strongly activated immediately after acute injuries like trauma, burn, or sepsis and helps to initiate regeneration. However, uncontrolled complement activation contributes to disease progression instead of supporting healing. Such effects are perceptible not only at the site of injury but also systemically, leading to systemic activation of other intravascular cascade systems eventually causing dysfunction of several vital organs. Understanding the complement pathomechanism and its interplay with other systems is a strict requirement for exploring novel therapeutic intervention routes. Ex vivo models exploring the cross-talk with other systems are rather limited, which complicates the determination of the exact pathophysiological roles that complement has in trauma, burn, and sepsis. Literature reporting on these three conditions is often controversial regarding the importance, distribution, and temporal occurrence of complement activation products further hampering the deduction of defined pathophysiological pathways driven by complement. Nevertheless, many in vitro experiments and animal models have shown beneficial effects of complement inhibition at different levels of the cascade. In the future, not only inhibition but also a complement reconstitution therapy should be considered in prospective studies to expedite how meaningful complement-targeted interventions need to be tailored to prevent complement augmented multi-organ failure after trauma, burn, and sepsis. This review summarizes clinically relevant studies investigating the role of complement in the acute diseases trauma, burn, and sepsis with important implications for clinical translation.
Collapse
Affiliation(s)
- Marco Mannes
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Helmholtzstr. 8/2, 89081, Ulm, Germany
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden.,Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Helmholtzstr. 8/2, 89081, Ulm, Germany.
| |
Collapse
|
14
|
Volovat SR, Negru S, Stolniceanu CR, Volovat C, Lungulescu C, Scripcariu D, Cobzeanu BM, Stefanescu C, Grigorescu C, Augustin I, Lupascu Ursulescu C, Volovat CC. Nanomedicine to modulate immunotherapy in cutaneous melanoma (Review). Exp Ther Med 2021; 21:535. [PMID: 33815608 PMCID: PMC8014970 DOI: 10.3892/etm.2021.9967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shifted the paradigm in cancer treatment in recent years. Immune checkpoint blockage (ICB), the active cancer vaccination and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer represent the main developments, achieving a surprising increased survival in patients included in clinical trials. In spite of these results, the current state-of-the-art immunotherapy has its limitations in efficacy. The existence of an interdisciplinary interface involving current knowledge in biology, immunology, bioengineering and materials science represents important progress in increasing the effectiveness of immunotherapy in cancer. Cutaneous melanoma remains a difficult cancer to treat, in which immunotherapy is a major therapeutic option. In fact, enhancing immunotherapy is possible using sophisticated biomedical nanotechnology platforms of organic or inorganic materials or engineering various immune cells to enhance the immune system. In addition, biological devices have developed, changing the approach to and treatment results in melanoma. In this review, we present different modalities to modulate the immune system, as well as opportunities and challenges in melanoma treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Serban Negru
- Department of Medical Oncology, 'Victor Babes' University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Dragos Scripcariu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Mihail Cobzeanu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Grigorescu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Corina Lupascu Ursulescu
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Radiology, 'Sf. Spiridon' Emergency Clinic Hospital, 700111 Iasi, Romania
| |
Collapse
|
15
|
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
|
16
|
Feng S, Zhang Z, Mo Y, Tong R, Zhong Z, Chen Z, He D, Wan R, Gao M, Mo Y, Zhang Q, Huang Y. Activation of NLRP3 inflammasome in hepatocytes after exposure to cobalt nanoparticles: The role of oxidative stress. Toxicol In Vitro 2020; 69:104967. [PMID: 32805375 DOI: 10.1016/j.tiv.2020.104967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/21/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022]
Abstract
With the increased use of nanomaterials and increased exposure of humans to various nanomaterials, the potential health effects of nanomaterials cannot be ignored. The hepatotoxicity of cobalt nanoparticles (Nano-Co) is largely unknown and the underlying mechanisms remain obscure. The purpose of this study was to exam the hepatotoxicity induced by Nano-Co and its potential mechanisms. Our results showed that exposure of human fetal hepatocytes L02 to Nano-Co caused a dose- and a time-dependent cytotoxicity. Besides the generation of reactive oxygen species (ROS) and mitochondrial reactive oxygen species (mtROS), exposure to Nano-Co also caused activation of NOD-like receptor protein 3 (NLRP3) inflammasome in hepatocytes. After silencing NLRP3, one component of NLRP3 inflammasome, expression by siRNA strategy, we found that upregulation of NLRP3-related proteins was abolished in hepatocytes exposed to Nano-Co. Using antioxidants to scavenge ROS and mtROS, we demonstrated that Nano-Co-induced mtROS generation was related to Nano-Co-induced NLRP3 inflammasome activation. Our findings demonstrated that Nano-Co exposure may promote intracellular oxidative stress damage, and mtROS may mediate the activation of NLRP3 inflammasome in hepatocytes exposed to Nano-Co, suggesting an important role of ROS/NLRP3 pathway in Nano-Co-induced hepatotoxicity. These results provide scientific insights into the hepatotoxicity of Nano-Co and a basis for the prevention and treatment of Nano-Co-induced cytotoxicity.
Collapse
Affiliation(s)
- Sisi Feng
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zhenyu Zhang
- Department of Emergency, Xiang'An Hospital of Xiamen University, Xiamen, Fujian, PR China
| | - Yiqing Mo
- Community Health Care Center, Changqing Chaoming Street, Xiacheng District, Hangzhou, Zhejiang, PR China
| | - Ruirui Tong
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zexiang Zhong
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zhong Chen
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Dan He
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Rong Wan
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Meiqin Gao
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yang Huang
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China.
| |
Collapse
|
17
|
Kargl R, Kleinschek KS. How can we understand the influence of nanoparticles on the coagulation of blood? Nanomedicine (Lond) 2020; 15:1923-1926. [PMID: 32677508 DOI: 10.2217/nnm-2020-0177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Rupert Kargl
- Institute for Chemistry & Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, Graz, 8010, Austria.,Institute for Automation, Faculty of Electrical Engineering & Computer Science, University of Maribor, Smetanova Ulica 17, Maribor, 2000, Slovenia
| | - Karin Stana Kleinschek
- Institute for Chemistry & Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, Graz, 8010, Austria.,Institute for Automation, Faculty of Electrical Engineering & Computer Science, University of Maribor, Smetanova Ulica 17, Maribor, 2000, Slovenia
| |
Collapse
|
18
|
Maisha N, Coombs T, Lavik E. Development of a Sensitive Assay to Screen Nanoparticles in vitro for Complement Activation. ACS Biomater Sci Eng 2020; 6:4903-4915. [PMID: 33313396 DOI: 10.1021/acsbiomaterials.0c00722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nanomedicines are often recognized by the innate immune system as a threat, leading to unwanted clearance due to complement activation. This adverse reaction not only alters the bioavailability of the therapeutic but can also cause cardiopulmonary complications and death in a portion of the population. There is a need for tools for assessing complement response in the early stage of development of nanomedicines. Currently, quantifying complement-mediated response in vitro is limited due to differences between in vitro and in vivo responses for the same precursors, differences in the complement systems in different species, and lack of highly sensitive tools for quantifying the changes. Hence, we have worked on developing complement assay conditions and sample preparation techniques that can be highly sensitive in assessing the complement-mediated response in vitro mimicking the in vivo activity. We are screening the impact of incubation time, nanoparticle dosage, anticoagulants, and species of the donor in both blood and blood components. We have validated the optimal assay conditions by replicating the impact of zeta potential seen in vivo on complement activation in vitro. As observed in our previous in vivo studies, where nanoparticles with neutral zeta-potential were able to suppress complement response, the change in the complement biomarker was least for the neutral nanoparticles as well through our developed guidelines. These assay conditions provide a vital tool for assessing the safety of intravenously administered nanomedicines.
Collapse
Affiliation(s)
- Nuzhat Maisha
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, Piscataway Territories
| | - Tobias Coombs
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, Piscataway Territories
| | - Erin Lavik
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, Piscataway Territories
| |
Collapse
|
19
|
Hobson JJ, Rannard SP, Owen A, Liptrott NJ. Safety assessment of a new nanoemulsion-based drug-delivery system reveals unexpected drug-free anticoagulant activity. Nanomedicine (Lond) 2020; 15:1361-1373. [PMID: 32484393 DOI: 10.2217/nnm-2019-0447] [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: A preclinical safety assessment of a novel nanoemulsion drug-delivery system, initially developed to improve the posology of efavirenz (EFV), was conducted with a specific focus on possible immunological and hematological complications. Materials & methods: Assessment of common acute toxicities, such as complement activation and cytokine secretion, was performed using validated assays known to have good correlation with in vivo end points. Results & conclusion: Compared with a standard aqueous solution of EFV, the EFV nanoemulsion showed no significant effect on immune cell function or phenotype. Prolongation of activated partial thromboplastin time was observed for EFV-loaded nanoemulsions (88% at 4 μg/ml) as well as unloaded nanoemulsions (52%) highlighting the potential for drug-free anticoagulant activity and warranting further investigation of the mechanism and utility of these materials.
Collapse
Affiliation(s)
- James J Hobson
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD, UK
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD, UK
| | - Andrew Owen
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GF, UK.,European Nanomedicine Characterisation Laboratory, University of Liverpool, Liverpool, L7 3NY, UK
| | - Neill J Liptrott
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GF, UK.,European Nanomedicine Characterisation Laboratory, University of Liverpool, Liverpool, L7 3NY, UK
| |
Collapse
|
20
|
Liu T, Bai R, Zhou H, Wang R, Liu J, Zhao Y, Chen C. The effect of size and surface ligands of iron oxide nanoparticles on blood compatibility. RSC Adv 2020; 10:7559-7569. [PMID: 35492144 PMCID: PMC9049842 DOI: 10.1039/c9ra10969b] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/11/2020] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely used and have attracted increased attention for their unique physicochemical properties, especially in biomedical sciences as contrast agents following intravenous administration. However, only few studies have systematically reported the blood compatibility of iron oxide nanoparticles with different physicochemical properties such as different sizes and surface ligands. Therefore, we selected three widely used organic ligands (polyacrylic acid, hyaluronic acid, and chitosan) with modified SPIONs at the same size of 5-6 nm, and polyacrylic acid-modified SPIONs with different sizes (5, 10, and 30 nm) at different concentrations to evaluate their haemocompatibility. Our results revealed that SPIONs modified with polyacrylic acid demonstrated size-dependent destruction of red blood cells and complement activation. Interestingly, 5 nm SPIONs prolonged blood clotting time as compared with 10 nm and 30 nm SPIONs in vitro. Compared with polyacrylic acid-modified SPIONs, hyaluronic acid- and chitosan-modified SPIONs least affected red blood cells, platelets, coagulation, and complement activation. Hence, hyaluronic acid- and chitosan-coated SPIONs are more suitable for nanomedicine applications than polyacrylic acid-coated SPIONs. Furthermore, the interaction between SPIONs and blood components strongly correlated with the administered concentration of nanoparticles. These results will provide some experimental information for safe-by-design SPIONs.
Collapse
Affiliation(s)
- Tao Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
| | - Rongqi Wang
- Department of Clinical Laboratory, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital Beijing 100080 P. R. China
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- Faculty of Life Sciences & Medicine, Northwest University (NWU) Xi'an 710069 P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. 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 Beijing 100190 P. R. China +86-10-62656765 +86 10 8254 5560
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
21
|
Mikelez-Alonso I, Aires A, Cortajarena AL. Cancer Nano-Immunotherapy from the Injection to the Target: The Role of Protein Corona. Int J Mol Sci 2020; 21:E519. [PMID: 31947622 PMCID: PMC7014289 DOI: 10.3390/ijms21020519] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has become a promising cancer therapy, improving the prognosis of patients with many different types of cancer and offering the possibility for long-term cancer remission. Nevertheless, some patients do not respond to these treatments and immunotherapy has shown some limitations, such as immune system resistance or limited bioavailability of the drug. Therefore, new strategies that include the use of nanoparticles (NPs) are emerging to enhance the efficacy of immunotherapies. NPs present very different pharmacokinetic and pharmacodynamic properties compared with free drugs and enable the use of lower doses of immune-stimulating molecules, minimizing their side effects. However, NPs face issues concerning stability in physiological conditions, protein corona (PC) formation, and accumulation in the target tissue. PC formation changes the physicochemical and biological properties of the NPs and in consequence their therapeutic effect. This review summarizes the recent advances in the study of the effects of PC formation in NP-based immunotherapy. PC formation has complex effects on immunotherapy since it can diminish ("immune blinding") or enhance the immune response in an uncontrolled manner ("immune reactivity"). Here, future perspectives of the field including the latest advances towards the use of personalized protein corona in cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Idoia Mikelez-Alonso
- CIC biomaGUNE, Parque Científico y Tecnológico de Gipuzkoa. Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain; (I.M.-A.); (A.A.)
- Immunopathology, BiocrucesBizkaia, Cruces Plaza, 48903 Barakaldo, Spain
| | - Antonio Aires
- CIC biomaGUNE, Parque Científico y Tecnológico de Gipuzkoa. Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain; (I.M.-A.); (A.A.)
| | - Aitziber L. Cortajarena
- CIC biomaGUNE, Parque Científico y Tecnológico de Gipuzkoa. Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain; (I.M.-A.); (A.A.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|