1
|
Liu L, Xu Y, Ma Y, Duan F, Wang C, Feng J, Yin H, Sun L, Li P, Li ZH. Fate of polystyrene micro- and nanoplastics in zebrafish liver cells: Influence of protein corona on transport, oxidative stress, and glycolipid metabolism. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137596. [PMID: 39952126 DOI: 10.1016/j.jhazmat.2025.137596] [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: 10/03/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Micro- and nanoplastics (MNPs) form protein corona (PC) upon contact with biological fluids, but their impact on the intracellular transport, distribution, and toxicity of MNPs remains unclear. Fetal bovine serum (FBS) and bovine serum albumin (BSA) were used to simulate in vivo environment, this study explored their influence on the transport and toxicity of polystyrene (PS) MNPs in zebrafish liver (ZFL) cells. Results showed PS MNPs were wrapped by proteins into stable complexes. Nanoparticles (NP, 50 nm) and their protein complexes (NP@PC) were internalized by cells within 6 h, with PC formation enhancing NP uptake. NP primarily entered cells through clathrin- and caveolae-mediated endocytosis, while NP@PC via clathrin-mediated pathways. Internalized particles were predominantly in lysosomes where PC degraded and some were also in mitochondria. Eventually, particles were expelled from cells through energy-dependent lysosomal pathways and energy-independent membrane penetration mechanisms. Notably, PC formation limited the clearance of NP. In toxicity, NP had a more severe impact than microplastics (MP, 5 μm). FBS more effectively mitigated PS MNPs-induced reactive oxygen species accumulation, subcellular structural damage, and dysregulation of glycolipid metabolism than BSA did. This study elucidates the modulatory role of PC on biological effects of MNPs, providing safety and risk management strategies.
Collapse
Affiliation(s)
- Ling Liu
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Yanan Xu
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Yuqing Ma
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Fengshang Duan
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Cunlong Wang
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Jianxue Feng
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Haiyang Yin
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Le Sun
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Ping Li
- Marine College, Shandong University, Shandong, Weihai 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Shandong, Weihai 264209, China.
| |
Collapse
|
2
|
Kocas M, Yamashita F, Comoglu T, Zhang Q. Enhancing Intracellular Uptake of Ivermectin through Liposomal Encapsulation. AAPS PharmSciTech 2025; 26:123. [PMID: 40316874 DOI: 10.1208/s12249-025-03113-8] [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: 11/19/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025] Open
Abstract
Ivermectin (IVM), an antiparasitic drug approved by the Food and Drug Administration (FDA), is widely used to treat several neglected tropical diseases, including onchocerciasis, helminthiases, and scabies. Additionally, IVM has shown potential as a potent inhibitor of certain RNA viruses, such as SARS-CoV-2. However, IVM is highly hydrophobic, essentially insoluble in water, which limits its bioavailability and therapeutic effectiveness. The use of liposomes as drug carriers offers several advantages, including enhanced solubility for lipophilic drugs, passive targeting of immune system cells, sustained release, and improved tissue penetration. To address the limitations of IVM, including its poor solubility and bioavailability, liposomal formulations were developed using a combination of soyphosphatidylcholine (SPC), dioleylphosphatidylcholine (DOPC), cholesterol (Ch), and diethylphosphate (DCP) in two distinct molar ratios (1.85:1:0.15 and 7:2:1) via the ethanol injection method. The physicochemical properties of the placebo and IVM-loaded liposomes were extensively characterized in our earlier study, including the particle size, polydispersity index, and zeta potential. The present work adds a deeper level of investigation into how to effect cellular uptake and cytotoxicity in vitro of both free IVM and IVM-loaded liposomes in Vero E6 cells. The half-maximal cytotoxic concentrations (CC50) for free IVM and IVM-loaded liposomes were 10 μM and > 110 μM, respectively and the cellular uptake of IVM-loaded liposomes ranged from 13 to 60%, whereas free IVM showed a significantly lower uptake of only 2%. These results demonstrate that liposomal encapsulation effectively enhances IVM's cellular uptake while reducing its cytotoxicity, thus offering a promising strategy for improving the effectiveness of IVM.
Collapse
Affiliation(s)
- Meryem Kocas
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Selcuk University, Selçuklu, Konya, 42130, Turkey
- Graduate School of Health Sciences, Ankara University, Dışkapı, Ankara, 06610, Turkey
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara University, Yenimahalle, Ankara, 06560, Turkey
- Graduate School of Pharmaceutical Sciences, Department of Drug Delivery Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Fumiyoshi Yamashita
- Graduate School of Pharmaceutical Sciences, Department of Quantitative Pharmaceutics, Kyoto University, Kyoto, 606-8501, Japan
- Graduate School of Pharmaceutical Sciences, Department of Drug Delivery Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Tansel Comoglu
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara University, Yenimahalle, Ankara, 06560, Turkey.
| | - Qiyue Zhang
- Graduate School of Pharmaceutical Sciences, Department of Quantitative Pharmaceutics, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
3
|
Szyk P, Czarczynska-Goslinska B, Ziegler-Borowska M, Larrosa I, Goslinski T. Sorafenib-Drug Delivery Strategies in Primary Liver Cancer. J Funct Biomater 2025; 16:148. [PMID: 40278256 PMCID: PMC12027913 DOI: 10.3390/jfb16040148] [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: 03/02/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Current primary liver cancer therapies, including sorafenib and transarterial chemoembolization, face significant limitations due to chemoresistance caused by impaired drug uptake, altered metabolism, and other genetic modulations. These challenges contribute to relapse rates of 50-80% within five years. The need for improved treatment strategies (adjuvant therapy, unsatisfactory enhanced permeability and retention (EPR) effect) has driven research into advanced drug delivery systems, including targeted nanoparticles, biomaterials, and combinatory approaches. Therefore, this review evaluates recent advancements in primary liver cancer pharmacotherapy, focusing on the potential of drug delivery systems for sorafenib and its derivatives. Approaches such as leveraging Kupffer cells for tumor migration or utilizing smaller NPs for inter-/intracellular delivery, address EPR limitations. Biomaterials and targeted therapies focusing on targeting have demonstrated effectiveness in increasing tumor-specific delivery, but clinical evidence remains limited. Combination therapies have emerged as an interesting solution to overcoming chemoresistance or to broadening therapeutic functionality. Biomimetic delivery systems, employing blood cells or exosomes, provide methods for targeting tumors, preventing metastasis, and strengthening immune responses. However, significant differences between preclinical models and human physiology remain a barrier to translating these findings into clinical success. Future research must focus on the development of adjuvant therapy and refining drug delivery systems to overcome the limitations of tumor heterogeneity and low drug accumulation.
Collapse
Affiliation(s)
- Piotr Szyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Beata Czarczynska-Goslinska
- Chair and Department of Pharmaceutical Technology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| | - Marta Ziegler-Borowska
- Department of Biomedical Chemistry and Polymer Science, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Igor Larrosa
- Department of Chemistry, University of Manchester, Chemistry Building, Oxford Road, Manchester M13 9PL, UK;
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
4
|
Ojansivu M, Barriga HMG, Holme MN, Morf S, Doutch JJ, Andaloussi SEL, Kjellman T, Johnsson M, Barauskas J, Stevens MM. Formulation and Characterization of Novel Ionizable and Cationic Lipid Nanoparticles for the Delivery of Splice-Switching Oligonucleotides. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419538. [PMID: 40091434 PMCID: PMC12038542 DOI: 10.1002/adma.202419538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/24/2025] [Indexed: 03/19/2025]
Abstract
Despite increasing knowledge about the mechanistic aspects of lipid nanoparticles (LNPs) as oligonucleotide carriers, the structure-function relationship in LNPs has been generally overlooked. Understanding this correlation is critical in the rational design of LNPs. Here, a materials characterization approach is utilized, applying structural information from small-angle X-ray scattering experiments to design novel LNPs focusing on distinct lipid organizations with a minimal compositional variation. The lipid phase structures are characterized in these LNPs and their corresponding bulk lipid mixtures with small-angle scattering techniques, and the LNP-cell interactions in vitro with respect to cytotoxicity, hemolysis, cargo delivery, cell uptake, and lysosomal swelling. An LNP is identified that outperforms Onpattro lipid composition using lipid components and molar ratios which differ from the gold standard clinical LNPs. The base structure of these LNPs has an inverse micellar phase organization, whereas the LNPs with inverted hexagonal phases are not functional, suggesting that this phase formation may not be needed for LNP-mediated oligonucleotide delivery. The importance of stabilizer choice for the LNP function is demonstrated and super-resolution microscopy highlights the complexity of the delivery mechanisms, where lysosomal swelling for the majority of LNPs is observed. This study highlights the importance of advanced characterization for the rational design of LNPs to enable the study of structure-function relationships.
Collapse
Affiliation(s)
- Miina Ojansivu
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - Hanna M. G. Barriga
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
- Present address:
Division of NanobiotechnologyDepartment of Protein ScienceSciLifeLab, KTH Royal Institute of TechnologySolnaSweden
| | - Margaret N. Holme
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - Stefanie Morf
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
| | - James J. Doutch
- ISIS Neutron and Muon SourceRutherford Appleton LaboratoryHarwell CampusOxfordshireOX11 0QXUK
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular MedicineDepartment of Laboratory MedicineKarolinska InstituteHuddinge14152StockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalStockholm141 86Sweden
- Karolinska ATMP CenterKarolinska InstituteHuddinge14152StockholmSweden
| | | | | | | | - Molly M. Stevens
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteHuddingeStockholm171 77Sweden
- Department of Physiology, Anatomy and GeneticsDepartment of Engineering ScienceKavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordOX1 3QUUK
| |
Collapse
|
5
|
Hammond J, Richards CJ, Ko Y, Jonker T, Åberg C, Roos WH, Lira RB. Membrane Fusion-Based Drug Delivery Liposomes Transiently Modify the Material Properties of Synthetic and Biological Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408039. [PMID: 40007088 PMCID: PMC11947515 DOI: 10.1002/smll.202408039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Many drug targets are located in intracellular compartments of cells but they often remain inaccessible to standard imaging and therapeutic agents. To aid intracellular delivery, drug carrier nanoparticles have been used to overcome the barrier imposed by the plasma membrane. The carrier must entrap large amounts of cargo, efficiently and quickly deliver the cargo in the cytosol or other intracellular compartments, and must be inert; they should not induce cellular responses or alter the cell state in the course of delivery. This study demonstrates that cationic liposomes with high charge density efficiently fuse with synthetic membranes and the plasma membrane of living cells. Direct fusion efficiently delivers large amounts of cargo to cells and cell-like vesicles within seconds, bypassing slow and often inefficient internalization-based pathways. These effects depend on liposome charge density, concentration, and the helper lipid. However, fusion-mediated cargo delivery results in the incorporation of large amounts of foreign lipids, causing changes to the material properties of these membranes, namely modifications in membrane packing and fluidity, induction of membrane curvature, decrease in surface tension, and the formation of (short-lived) pores. Importantly, these effects are transient and liposome removal allows cells to recover their state prior to liposome interaction.
Collapse
Affiliation(s)
- Jayna Hammond
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Ceri J. Richards
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - YouBeen Ko
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Thijs Jonker
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Christoffer Åberg
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - Wouter H. Roos
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Rafael B. Lira
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Present address:
Department of BionanoscienceKavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| |
Collapse
|
6
|
Araie H, Hosono N, Yamauchi T. Cellular uptake of CPX-351 by scavenger receptor class B type 1-mediated nonendocytic pathway. Exp Hematol 2024; 140:104651. [PMID: 39362576 DOI: 10.1016/j.exphem.2024.104651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
The proper uptake of drugs in liposome formulations into target cells markedly impacts therapeutic efficacy. The protein corona (PC), formed by the adsorption of serum proteins onto the liposome surface, binds to specific surface receptors of target cells, influencing the uptake pathway. We investigated the uptake pathway into leukemia cells based on PC analysis of CPX-351, a liposome containing cytarabine and daunorubicin in a fixed 5:1 synergistic molar ratio. The PC of CPX-351 mixed with fetal bovine serum was analyzed by nanoflow liquid chromatography-tandem mass spectrometry. CPX-351 uptake in HL-60, K562, and THP-1 leukemia cell lines was measured by flow cytometry using daunorubicin fluorescence. The major components of CPX-351 PC include apolipoproteins A-I and A-II, which bind to scavenger receptor class B type 1 (SR-BI), a nonendocytic pathway that takes up only liposome contents. SR-BI was expressed in each cell, and its expression correlated with CPX-351 uptake. The uptake was significantly decreased by the inhibition of clathrin-mediated endocytosis and macropinocytosis. Additionally, blocks lipid transport-1 (BLT-1), a selective inhibitor of SR-BI, decreased the uptake; however, high-dose BLT-1 addition significantly increased the uptake, which was more strongly inhibited by macropinocytosis suppression compared with clathrin-mediated endocytosis. BLT-1 enhances the binding of SR-BI to liposomes in a dose-dependent manner. These findings indicate that the enhancement of binding between SR-BI and CPX-351 activates different pathways, such as macropinocytosis, distinct from CPX-351 alone. SR-BI may be a biomarker for CPX-351 therapy, and the combination of CPX-351 with high-dose BLT-1 may augment therapeutic efficacy.
Collapse
Affiliation(s)
- Hiroaki Araie
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Naoko Hosono
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
7
|
Martin A, Zhang S, Williamson A, Tingley B, Pickus M, Zurakowski D, Nia HT, Shirihai O, Han X, Grinstaff MW. Universal high-throughput image quantification of subcellular structure dynamics and spatial distributions within cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.18.608451. [PMID: 39229224 PMCID: PMC11370428 DOI: 10.1101/2024.08.18.608451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Image analysis of subcellular structures and biological processes relies on specific, context-dependent pipelines, which are labor-intensive, constrained by the intricacies of the specific biological system, and inaccessible to broader applications. Here we introduce the application of dispersion indices, a statistical tool traditionally employed by economists, to analyze the spatial distribution and heterogeneity of subcellular structures. This computationally efficient high-throughput approach, termed GRID (Generalized Readout of Image Dispersion), is highly generalizable, compatible with open-source image analysis software, and adaptable to diverse biological scenarios. GRID readily quantifies diverse structures and processes to include autophagic puncta, mitochondrial clustering, and microtubule dynamics. Further, GRID is versatile, applicable to both 2D cell cultures and 3D multicellular organoids, and suitable for high-throughput screening and performance metric measurements, such as half-maximal effective concentration (EC50) values. The approach enables mechanistic analysis of critical subcellular structure processes of relevance for diseases ranging from metabolic and neuronal diseases to cancer as well as a first-pass screening method for identifying biologically active agents for drug discovery.
Collapse
Affiliation(s)
- Andrew Martin
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Amanda Williamson
- Department of Chemistry, Boston University, Boston, MA 02215, United States
| | - Brett Tingley
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Mira Pickus
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | | | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Orian Shirihai
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Mark W. Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
- Department of Chemistry, Boston University, Boston, MA 02215, United States
| |
Collapse
|
8
|
Mendes M, Nunes S, Cova T, Branco F, Dyrks M, Koksch B, Vale N, Sousa J, Pais A, Vitorino C. Charge-switchable cell-penetrating peptides for rerouting nanoparticles to glioblastoma treatment. Colloids Surf B Biointerfaces 2024; 241:113983. [PMID: 38850741 DOI: 10.1016/j.colsurfb.2024.113983] [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: 12/12/2023] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024]
Abstract
Glioblastoma (GB) is one of the most lethal types of neoplasms with unique anatomic, physiologic, and pathologic features that usually persist after exposure to standard therapeutic modalities. It is biologically aggressive, and the existence of the blood-brain barrier (BBB) limits the efficacy of standard therapies. In this work, we hypothesize the potential of surface-functionalized ultra-small nanostructured lipid carriers (usNLCs) with charge-switchable cell-penetrating peptides (CPPs) to overcome this biological barrier and improve targeted delivery to brain tumor tissues. The big question is: what is the potential of CPPs in directing nanoparticles toward brain tumor tissue? To answer this question, the usNLCs were functionalized with distinct biomolecules [five CPPs, c(RGDfK) and transferrin, Tf] through electrostatic interaction and its ability as a targeting approach to BBB (HBMEC) and glioma cells (U87 cells) evaluated in terms of physicochemical properties, cellular uptake, permeability in a 2D-BBB model, and tumor growth inhibition. Monte Carlo simulations elucidated CPP adsorption patterns. The permeability studies revealed that targeted usNLCs, especially usNLCsTf and usNLCsCPP4, exhibited an increased permeability coefficient compared to the non-targeted usNLCs. Functionalized usNLCs evidenced enhanced uptake in BBB cells, with smaller CPPs showing higher internalization (CPP1 and CPP2). Similarly, functionalized usNLCs exhibited more significant cytotoxicity in glioma cells, with specific CPPs promoting favorable internalization. Analysis of the endocytic pathway indicated that usNLCsCPPs were mainly internalized by direct translocation and caveolae-mediated endocytosis. Optimal usNLCs with dual targeting capabilities to both BBB and GB cells provide a promising therapeutic strategy for GB.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal
| | - Sandra Nunes
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal
| | - Tânia Cova
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal
| | - Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal
| | - Michael Dyrks
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Arnimallee 20, Berlin 14195, Germany
| | - Beate Koksch
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Arnimallee 20, Berlin 14195, Germany
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto (FFUP), Alameda Professor Hernâni Monteiro, Porto 4200-319, Portugal; Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, Porto 4200-450, Portugal
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra 3004-535, Portugal.
| |
Collapse
|
9
|
Caracciolo G. Artificial protein coronas: directing nanoparticles to targets. Trends Pharmacol Sci 2024; 45:602-613. [PMID: 38811308 DOI: 10.1016/j.tips.2024.05.003] [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: 03/30/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
The protein corona surrounding nanoparticles (NPs) offers exciting possibilities for targeted drug delivery. However, realizing this potential requires direct evidence of corona-receptor interactions in vivo; a challenge hampered by the limitations of in vitro settings. This opinion proposes that utilizing engineered protein coronas can address this challenge. Artificial coronas made of selected plasma proteins retain their properties in vivo, enabling manipulation for specific receptor targeting. To directly assess corona-receptor interactions mimicking in vivo complexity, we propose testing artificial coronas with recently adapted quartz crystal microbalance (QCM) setups whose current limitations and potential advancements are critically discussed. Finally, the opinion proposes future experiments to decipher corona-receptor interactions and unlock the full potential of the protein corona for NP-based drug delivery.
Collapse
Affiliation(s)
- Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
10
|
Giulimondi F, Digiacomo L, Renzi S, Cassone C, Pirrottina A, Molfetta R, Palamà IE, Maiorano G, Gigli G, Amenitsch H, Pozzi D, Zingoni A, Caracciolo G. Optimizing Transfection Efficiency in CAR-T Cell Manufacturing through Multiple Administrations of Lipid-Based Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3746-3757. [PMID: 38775109 DOI: 10.1021/acsabm.4c00103] [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] [Indexed: 06/18/2024]
Abstract
The existing manufacturing protocols for CAR-T cell therapies pose notable challenges, particularly in attaining a transient transfection that endures for a significant duration. To address this gap, this study aims to formulate a transfection protocol utilizing multiple lipid-based nanoparticles (LNPs) administrations to enhance transfection efficiency (TE) to clinically relevant levels. By systematically fine-tuning and optimizing our transfection protocol through a series of iterative refinements, we have accomplished a remarkable one-order-of-magnitude augmentation in TE within the immortalized T-lymphocyte Jurkat cell line. This enhancement has been consistently observed over 2 weeks, and importantly, it has been achieved without any detrimental impact on cell viability. In the subsequent phase of our study, we aimed to optimize the gene delivery system by evaluating three lipid-based formulations tailored for DNA encapsulation using our refined protocol. These formulations encompassed two LNPs constructed from ionizable lipids and featuring systematic variations in lipid composition (iLNPs) and a cationic lipoplex (cLNP). Our findings showcased a notable standout among the three formulations, with cLNP emerging as a frontrunner for further refinement and integration into the production pipeline of CAR-T therapies. Consequently, cLNP was scrutinized for its potential to deliver CAR-encoding plasmid DNA to the HEK-293 cell line. Confocal microscopy experiments demonstrated its efficiency, revealing substantial internalization compared to iLNPs. By employing a recently developed confocal image analysis method, we substantiated that cellular entry of cLNP predominantly occurs through macropinocytosis. This mechanism leads to heightened intracellular endosomal escape and mitigates lysosomal accumulation. The successful expression of anti-CD19-CD28-CD3z, a CAR engineered to target CD19, a protein often expressed on the surface of B cells, was confirmed using a fluorescence-based assay. Overall, our results indicated the effectiveness of cLNP in gene delivery and suggested the potential of multiple administration transfection as a practical approach for refining T-cell engineering protocols in CAR therapies. Future investigations may focus on refining outcomes by adjusting transfection parameters like nucleic acid concentration, lipid-to-DNA ratio, and incubation time to achieve improved TE and increased gene expression levels.
Collapse
Affiliation(s)
- Francesca Giulimondi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Serena Renzi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Chiara Cassone
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Andrea Pirrottina
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | | | - Gabriele Maiorano
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, Lecce 73100, Italy
| | - Giuseppe Gigli
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, Lecce 73100, Italy
- Department of Medicine, University of Salento, Arnesano street c/o Campus Ecotekne, Lecce 73100, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Graz 8010, Austria
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| |
Collapse
|
11
|
Gao Y, Huang Y, Ren C, Chou P, Wu C, Pan X, Quan G, Huang Z. Looking back, moving forward: protein corona of lipid nanoparticles. J Mater Chem B 2024; 12:5573-5588. [PMID: 38757190 DOI: 10.1039/d4tb00186a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Lipid nanoparticles (LNPs) are commonly employed for drug delivery owing to their considerable drug-loading capacity, low toxicity, and excellent biocompatibility. Nevertheless, the formation of protein corona (PC) on their surfaces significantly influences the drug's in vivo fate (such as absorption, distribution, metabolism, and elimination) upon administration. PC denotes the phenomenon wherein one or multiple strata of proteins adhere to the external interface of nanoparticles (NPs) or microparticles within the biological milieu, encompassing ex vivo fluids (e.g., serum-containing culture media) and in vivo fluids (such as blood and tissue fluids). Hence, it is essential to claim the PC formation behaviors and mechanisms on the surface of LNPs. This overview provided a comprehensive examination of crucial aspects related to such issues, encompassing time evolution, controllability, and their subsequent impacts on LNPs. Classical studies of PC generation on the surface of LNPs were additionally integrated, and its decisive role in shaping the in vivo fate of LNPs was explored. The mechanisms underlying PC formation, including the adsorption theory and alteration theory, were introduced to delve into the formation process. Subsequently, the existing experimental outcomes were synthesized to offer insights into the research and application facets of PC, and it was concluded that the manipulation of PC held substantial promise in the realm of targeted delivery.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Yeqi Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Chuanyu Ren
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Peiwen Chou
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| |
Collapse
|
12
|
Poinsot V, Pizzinat N, Ong-Meang V. Engineered and Mimicked Extracellular Nanovesicles for Therapeutic Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:639. [PMID: 38607173 PMCID: PMC11013861 DOI: 10.3390/nano14070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Exosomes are spherical extracellular nanovesicles with an endosomal origin and unilamellar lipid-bilayer structure with sizes ranging from 30 to 100 nm. They contain a large range of proteins, lipids, and nucleic acid species, depending on the state and origin of the extracellular vesicle (EV)-secreting cell. EVs' function is to encapsulate part of the EV-producing cell content, to transport it through biological fluids to a targeted recipient, and to deliver their cargos specifically within the aimed recipient cells. Therefore, exosomes are considered to be potential biological drug-delivery systems that can stably deliver their cargo into targeted cells. Various cell-derived exosomes are produced for medical issues, but their use for therapeutic purposes still faces several problems. Some of these difficulties can be avoided by resorting to hemisynthetic approaches. We highlight here the uses of alternative exosome-mimes involving cell-membrane coatings on artificial nanocarriers or the hybridization between exosomes and liposomes. We also detail the drug-loading strategies deployed to make them drug-carrier systems and summarize the ongoing clinical trials involving exosomes or exosome-like structures. Finally, we summarize the open questions before considering exosome-like disposals for confident therapeutic delivery.
Collapse
Affiliation(s)
- Verena Poinsot
- Inserm, CNRS, Faculté de Santé, Université Toulouse III—Paul Sabatier, I2MC U1297, 31432 Toulouse, France; (N.P.); (V.O.-M.)
| | | | | |
Collapse
|
13
|
Muenzebrock KA, Ho FYW, Pontes AP, Jorquera-Cordero C, Utomo L, Garcia JP, Willems PC, Welting TJM, Rip J, Creemers LB. Polymeric Nanoparticles Enable mRNA Transfection and Its Translation in Intervertebral Disc and Human Joint Cells, Except for M1 Macrophages. Pharmaceutics 2024; 16:438. [PMID: 38675100 PMCID: PMC11053495 DOI: 10.3390/pharmaceutics16040438] [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: 02/20/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic lower back pain caused by intervertebral disc degeneration and osteoarthritis (OA) are highly prevalent chronic diseases. Although pain management and surgery can alleviate symptoms, no disease-modifying treatments are available. mRNA delivery could halt inflammation and degeneration and induce regeneration by overexpressing anti-inflammatory cytokines or growth factors involved in cartilage regeneration. Here, we investigated poly(amidoamine)-based polymeric nanoparticles to deliver mRNA to human joint and intervertebral disc cells. Human OA chondrocytes, human nucleus pulposus (NP) cells, human annulus fibrosus (AF) cells, fibroblast-like synoviocytes (FLS) and M1-like macrophages were cultured and transfected with uncoated or PGA-PEG-coated nanoparticles loaded with EGFP-encoding mRNA. Cell viability and transfection efficiency were analyzed for all cell types. Nanoparticle internalization was investigated in FLS and M1-like macrophages. No significant decrease in cell viability was observed in most conditions. Only macrophages showed a dose-dependent reduction of viability. Transfection with either nanoparticle version resulted in EGFP expression in NP cells, AF cells, OA chondrocytes and FLS. Macrophages showed internalization of nanoparticles by particle-cell co-localization, but no detectable expression of EGFP. Taken together, our data show that poly (amidoamine)-based nanoparticles can be used for mRNA delivery into cells of the human joint and intervertebral disc, indicating its potential future use as an mRNA delivery system in OA and IVDD, except for macrophages.
Collapse
Affiliation(s)
- Katrin Agnes Muenzebrock
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (K.A.M.)
| | - Fiona Y. W. Ho
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (K.A.M.)
| | - Adriano P. Pontes
- 20Med Therapeutics BV, 2333 BD Leiden, The Netherlands; (A.P.P.); (J.R.)
| | - Carla Jorquera-Cordero
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Lizette Utomo
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Joao Pedro Garcia
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (K.A.M.)
| | - Paul C. Willems
- Department of Orthopedic Surgery, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (P.C.W.); (T.J.M.W.)
| | - Tim J. M. Welting
- Department of Orthopedic Surgery, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (P.C.W.); (T.J.M.W.)
| | - Jaap Rip
- 20Med Therapeutics BV, 2333 BD Leiden, The Netherlands; (A.P.P.); (J.R.)
| | - Laura B. Creemers
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (K.A.M.)
| |
Collapse
|
14
|
Kimura S, Harashima H. Nano-Bio Interactions: Exploring the Biological Behavior and the Fate of Lipid-Based Gene Delivery Systems. BioDrugs 2024; 38:259-273. [PMID: 38345754 DOI: 10.1007/s40259-024-00647-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 03/06/2024]
Abstract
Gene therapy for many diseases is rapidly becoming a reality, as demonstrated by the recent approval of various nucleic acid-based therapeutics. Non-viral systems such as lipid-based carriers, lipid nanoparticles (LNPs), for delivering different payloads including small interfering RNA, plasmid DNA, and messenger RNA have been particularly extensively explored and developed for clinical uses. One of the most important issues in LNP development is delivery to extrahepatic tissues. To achieve this, various lipids and lipid-like materials are being examined and screened. Several LNP formulations that target extrahepatic tissues, such as the spleen and the lungs have been developed by adjusting the lipid compositions of LNPs. However, mechanistic details of how the characteristics of LNPs affect delivery efficiency remains unclear. The purpose of this review is to provide an overview of LNP-based nucleic acid delivery focusing on LNP components and their structures, as well as discussing biological factors, such as biomolecular corona and cellular responses related to the delivery efficiency.
Collapse
Affiliation(s)
- Seigo Kimura
- Integrated Research Consortium on Chemical Sciences, Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan.
| | - Hideyoshi Harashima
- Laboratory for Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
15
|
Neal CJ, Kolanthai E, Wei F, Coathup M, Seal S. Surface Chemistry of Biologically Active Reducible Oxide Nanozymes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2211261. [PMID: 37000888 DOI: 10.1002/adma.202211261] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/15/2023] [Indexed: 06/19/2023]
Abstract
Reducible metal oxide nanozymes (rNZs) are a subject of intense recent interest due to their catalytic nature, ease of synthesis, and complex surface character. Such materials contain surface sites which facilitate enzyme-mimetic reactions via substrate coordination and redox cycling. Further, these surface reactive sites are shown to be highly sensitive to stresses within the nanomaterial lattice, the physicochemical environment, and to processing conditions occurring as part of their syntheses. When administered in vivo, a complex protein corona binds to the surface, redefining its biological identity and subsequent interactions within the biological system. Catalytic activities of rNZs each deliver a differing impact on protein corona formation, its composition, and in turn, their recognition, and internalization by host cells. Improving the understanding of the precise principles that dominate rNZ surface-biomolecule adsorption raises the question of whether designer rNZs can be engineered to prevent corona formation, or indeed to produce "custom" protein coronas applied either in vitro, and preadministration, or formed immediately upon their exposure to body fluids. Here, fundamental surface chemistry processes and their implications in rNZ material performance are considered. In particular, material structures which inform component adsorption from the application environment, including substrates for enzyme-mimetic reactions are discussed.
Collapse
Affiliation(s)
- Craig J Neal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Fei Wei
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Melanie Coathup
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
16
|
Guo F, Luo S, Wang L, Wang M, Wu F, Wang Y, Jiao Y, Du Y, Yang Q, Yang X, Yang G. Protein corona, influence on drug delivery system and its improvement strategy: A review. Int J Biol Macromol 2024; 256:128513. [PMID: 38040159 DOI: 10.1016/j.ijbiomac.2023.128513] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Nano drug delivery systems offer several benefits, including enhancing drug solubility, regulating drug release, prolonging drug circulation time, and minimized toxicity and side effects. However, upon entering the bloodstream, nanoparticles (NPs) encounter a complex biological environment and get absorbed by various biological components, primarily proteins, leading to the formation of a 'Protein Corona'. The formation of the protein corona is affected by the characteristics of NPs, the physiological environment, and experimental design, which in turn affects of the immunotoxicity, specific recognition, cell uptake, and drug release of NPs. To improve the abundance of a specific protein on NPs, researchers have explored pre-coating, modifying, or wrapping NPs with the cell membrane to reduce protein adsorption. This paper, we have reviewed studies of the protein corona in recent years, summarized the formation and detection methods of the protein corona, the effect of the protein corona composition on the fate of NPs, and the design of new drug delivery systems based on the optimization of protein corona to provide a reference for further study of the protein corona and a theoretical basis for the clinical transformation of NPs.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shuai Luo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lianyi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengqi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fang Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujia Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yunlong Jiao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yinzhou Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qingliang Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaoyan Yang
- Zhejiang Provincial People's Hospital, Hangzhou 314408, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
17
|
Dietz L, Oberländer J, Mateos‐Maroto A, Schunke J, Fichter M, Krämer‐Albers E, Landfester K, Mailänder V. Uptake of extracellular vesicles into immune cells is enhanced by the protein corona. J Extracell Vesicles 2023; 12:e12399. [PMID: 38124271 PMCID: PMC10733601 DOI: 10.1002/jev2.12399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
The influence of a protein corona on the uptake of nanoparticles in cells has been demonstrated in various publications over the last years. Extracellular vesicles (EVs), can be seen as natural nanoparticles. However, EVs are produced under different cell culture conditions and little is known about the protein corona forming on EVs and its influence on their uptake by target cells. Here, we use a proteomic approach in order to analyze the protein composition of the EVs themselves and the protein composition of a human blood plasma protein corona around EVs. Moreover, we analyze the influence of the protein corona on EV uptake into human monocytes and compare it with the influence on the uptake of engineered liposomes. We show that the presence of a protein corona increases the uptake of EVs in human monocytes. While for liposomes this seems to be triggered by the presence of immunoglobulins in the protein corona, for EVs blocking the Fc receptors on monocytes did not show an influence of uptake. Therefore, other mechanisms of docking to the cell membrane and uptake are most like involved, demonstrating a clear difference between EVs and liposomes as technically produced nanocarriers.
Collapse
Affiliation(s)
- Laura Dietz
- Department of DermatologyUniversity Medical Center MainzMainzGermany
- Max Planck Institute for Polymer ResearchMainzGermany
| | - Jennifer Oberländer
- Department of DermatologyUniversity Medical Center MainzMainzGermany
- Max Planck Institute for Polymer ResearchMainzGermany
| | | | - Jenny Schunke
- Department of DermatologyUniversity Medical Center MainzMainzGermany
- Max Planck Institute for Polymer ResearchMainzGermany
| | - Michael Fichter
- Department of DermatologyUniversity Medical Center MainzMainzGermany
- Max Planck Institute for Polymer ResearchMainzGermany
| | - Eva‐Maria Krämer‐Albers
- Institute of Developmental Biology and NeurobiologyJohannes Gutenberg University of MainzMainzGermany
| | | | - Volker Mailänder
- Department of DermatologyUniversity Medical Center MainzMainzGermany
- Max Planck Institute for Polymer ResearchMainzGermany
| |
Collapse
|
18
|
Pozzi D, Caracciolo G. Looking Back, Moving Forward: Lipid Nanoparticles as a Promising Frontier in Gene Delivery. ACS Pharmacol Transl Sci 2023; 6:1561-1573. [PMID: 37974625 PMCID: PMC10644400 DOI: 10.1021/acsptsci.3c00185] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Indexed: 11/19/2023]
Abstract
Lipid nanoparticles (LNPs) have shown remarkable success in delivering genetic materials like COVID-19 LNP vaccines, such as mRNA-1273/SpikeVax by Moderna and BNT162b2/Comirnaty by BioNTech/Pfizer, as well as siRNA for rare inherited diseases, such as Onpattro from Alnylam Pharmaceuticals. These LNPs are advantageous since they minimize side effects, target specific cells, and regulate payload delivery. There has been a surge of interest in these particles due to their success stories; however, we still do not know much about how they work. This perspective will recapitulate the evolution of lipid-based gene delivery, starting with Felgner's pioneering 1987 PNAS paper, which introduced the initial DNA-transfection method utilizing a synthetic cationic lipid. Our journey takes us to the early 2020s, a time when advancements in bionano interactions enabled us to create biomimetic lipoplexes characterized by a remarkable ability to evade capture by immune cells in vivo. Through this overview, we propose leveraging previous achievements to assist us in formulating improved research goals when optimizing LNPs for medical conditions such as infectious diseases, cancer, and heritable disorders.
Collapse
Affiliation(s)
- Daniela Pozzi
- NanoDelivery Lab, Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina
Elena 291, 00161 Rome, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina
Elena 291, 00161 Rome, Italy
| |
Collapse
|
19
|
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
|
20
|
Kimura S, Harashima H. On the mechanism of tissue-selective gene delivery by lipid nanoparticles. J Control Release 2023; 362:797-811. [PMID: 37004796 DOI: 10.1016/j.jconrel.2023.03.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023]
Abstract
The era of nucleic acid nanomedicine has arrived, as evidenced by Patisiran, a small interfering RNA (siRNA) encapsulated lipid nanoparticle (LNP), and mRNA-loaded LNPs used in COVID-19 vaccines. The diversity of nano-designs for delivering nucleic acid molecules tested in Phase II/III clinical trials reflects the potential of these technologies. These breakthroughs in non-viral gene delivery, including the use of LNPs, have attracted substantial interest worldwide for developing more effective drugs. A next step in this field is to target tissues other than the liver, which requires significant research efforts and material development. However, mechanistic studies in this area are lacking. This study compares two types of LNPs with different tissue-selectivity for delivering plasmid DNA (pDNA), one being liver-selective and the other spleen-selective, in an effort to understand the mechanisms responsible for differences in gene expression of delivered genes. We observed little difference in the biodistribution of these two LNPs despite the 100-1000-fold differences in gene expression. We then quantified the amount of delivered pDNA and mRNA expression in each tissue by quantitative real-time PCR (qPCR) to evaluate various intracellular processes, such as nuclear delivery, transcription and translation. The results showed a >100-fold difference in the translation step but there were little differences in amount of pDNA delivered to the nucleus or the amount of mRNA expression for the two LNP deliveries. Our findings suggest that endogenous factors affect gene expression efficiency not the extent of biodistribution.
Collapse
Affiliation(s)
- Seigo Kimura
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
21
|
Gandek TB, van der Koog L, Nagelkerke A. A Comparison of Cellular Uptake Mechanisms, Delivery Efficacy, and Intracellular Fate between Liposomes and Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2300319. [PMID: 37384827 PMCID: PMC11469107 DOI: 10.1002/adhm.202300319] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
A key aspect for successful drug delivery via lipid-based nanoparticles is their internalization in target cells. Two prominent examples of such drug delivery systems are artificial phospholipid-based carriers, such as liposomes, and their biological counterparts, the extracellular vesicles (EVs). Despite a wealth of literature, it remains unclear which mechanisms precisely orchestrate nanoparticle-mediated cargo delivery to recipient cells and the subsequent intracellular fate of therapeutic cargo. In this review, internalization mechanisms involved in the uptake of liposomes and EVs by recipient cells are evaluated, also exploring their intracellular fate after intracellular trafficking. Opportunities are highlighted to tweak these internalization mechanisms and intracellular fates to enhance the therapeutic efficacy of these drug delivery systems. Overall, literature to date shows that both liposomes and EVs are predominantly internalized through classical endocytosis mechanisms, sharing a common fate: accumulation inside lysosomes. Studies tackling the differences between liposomes and EVs, with respect to cellular uptake, intracellular delivery and therapy efficacy, remain scarce, despite its importance for the selection of an appropriate drug delivery system. In addition, further exploration of functionalization strategies of both liposomes and EVs represents an important avenue to pursue in order to control internalization and fate, thereby improving therapeutic efficacy.
Collapse
Affiliation(s)
- Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
22
|
Mao X, Wang G, Wang Z, Duan C, Wu X, Xu H. Theranostic Lipid Nanoparticles for Renal Cell Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2306246. [PMID: 37747365 DOI: 10.1002/adma.202306246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Renal cell carcinoma (RCC) is a common urological malignancy and represents a leading threat to healthcare. Recent years have seen a series of progresses in the early diagnosis and management of RCC. Theranostic lipid nanoparticles (LNPs) are increasingly becoming one of the focuses in this field, because of their suitability for tumor targeting and multimodal therapy. LNPs can be precisely fabricated with desirable chemical compositions and biomedical properties, which closely match the physiological characteristics and clinical needs of RCC. Herein, a comprehensive review of theranostic LNPs is presented, emphasizing the generic tool nature of LNPs in developing advanced micro-nano biomaterials. It begins with a brief overview of the compositions and formation mechanism of LNPs, followed with an introduction to kidney-targeting approaches, such as passive, active, and stimulus responsive targeting. With examples provided, a series of modification strategies for enhancing the tumor targeting and functionality of LNPs are discussed. Thereafter, research advances on applications of these LNPs for RCC including bioimaging, liquid biopsy, drug delivery, physical therapy, and gene therapy are summarized and discussed from an interdisciplinary perspective. The final part highlights the milestone achievements of translation medicine, current challenges as well as future development directions of LNPs for the diagnosis and treatment of RCC.
Collapse
Affiliation(s)
- Xiongmin Mao
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guanyi Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zijian Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chen Duan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoliang Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hua Xu
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
23
|
Tretiakova D, Kobanenko M, Alekseeva A, Boldyrev I, Khaidukov S, Zgoda V, Tikhonova O, Vodovozova E, Onishchenko N. Protein Corona of Anionic Fluid-Phase Liposomes Compromises Their Integrity Rather than Uptake by Cells. MEMBRANES 2023; 13:681. [PMID: 37505047 PMCID: PMC10384875 DOI: 10.3390/membranes13070681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/27/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023]
Abstract
Despite the undisputable role of the protein corona in the biointeractions of liposome drug carriers, the field suffers from a lack of knowledge regarding the patterns of protein deposition on lipid surfaces with different compositions. Here, we investigated the protein coronas formed on liposomes of basic compositions containing combinations of egg phosphatidylcholine (PC), palmitoyloleoyl phosphatidylglycerol (POPG), and cholesterol. Liposome-protein complexes isolated by size-exclusion chromatography were delipidated and analyzed using label-free LC-MS/MS. The addition of the anionic lipid and cholesterol both affected the relative protein abundances (and not the total bound proteins) in the coronas. Highly anionic liposomes, namely those containing 40% POPG, carried corona enriched with cationic proteins (apolipoprotein C1, beta-2-glycoprotein 1, and cathelicidins) and were the least stable in the calcein release assay. Cholesterol improved the liposome stability in the plasma. However, the differences in the corona compositions had little effect on the liposome uptake by endothelial (EA.hy926) and phagocytic cells in the culture (U937) or ex vivo (blood-derived monocytes and neutrophils). The findings emphasize that the effect of protein corona on the performance of the liposomes as drug carriers occurs through compromising particle stability rather than interfering with cellular uptake.
Collapse
Affiliation(s)
- Daria Tretiakova
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria Kobanenko
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Anna Alekseeva
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Ivan Boldyrev
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Sergey Khaidukov
- Laboratory of Carbohydrates, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Viktor Zgoda
- Institute of Biomedical Chemistry, ul. Pogodinskaya 10, 119121 Moscow, Russia
| | - Olga Tikhonova
- Institute of Biomedical Chemistry, ul. Pogodinskaya 10, 119121 Moscow, Russia
| | - Elena Vodovozova
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Natalia Onishchenko
- Laboratory of Lipid Chemistry, Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
24
|
Mateos-Maroto A, Gai M, Brückner M, da Costa Marques R, Harley I, Simon J, Mailänder V, Morsbach S, Landfester K. Systematic modulation of the lipid composition enables the tuning of liposome cellular uptake. Acta Biomater 2023; 158:463-474. [PMID: 36599401 DOI: 10.1016/j.actbio.2022.12.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023]
Abstract
As liposomes have been widely explored as drug delivery carriers over the past decades, they are one of the most promising platforms due to their biocompatibility and versatility for surface functionalization. However, to improve the specific design of liposomes for future biomedical applications such as nanovaccines, it is necessary to understand how these systems interact with cell membranes, as most of their potential applications require them to be internalized by cells. Even though several investigations on the cellular uptake of liposomes were conducted, the effect of the liposome membrane properties on internalization in different cell lines remains unclear. Here, we demonstrate how the cellular uptake behavior of liposomes can be driven towards preferential interaction with dendritic cells (DC2.4) as compared to macrophages (RAW264.7) by tuning the lipid composition with varied molar ratios of the lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). Cellular internalization efficiency was analyzed by flow cytometry, as well as liposome-cell membrane co-localization by confocal laser scanning microscopy. The corresponding proteomic analysis of the protein corona was performed in order to unravel the possible effect on the internalization. The obtained results of this work reveal that it is possible to modulate the cellular uptake towards enhanced internalization by dendritic cells just by modifying the applied lipids and, thus, mainly the physico-chemical properties of the liposomes. STATEMENT OF SIGNIFICANCE: In the field of nanomedicine, it is of key importance to develop new specific and efficient drug carriers. In this sense, liposomes are one of the most widely known carrier types and used in clinics with good results. However, the exact interaction mechanisms of liposomes with cells remain unclear, which is of great importance for the design of new drug delivery platforms. Therefore, in this work we demonstrate that cellular uptake depends on the lipid composition. We are able to enhance the uptake in a specific cell type just by tuning the content of a lipid in the liposome membrane. This finding could be a step towards the selective design of liposomes to be internalized by specific cells with promising applications in biomedicine.
Collapse
Affiliation(s)
- Ana Mateos-Maroto
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Meiyu Gai
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Maximilian Brückner
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Richard da Costa Marques
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Iain Harley
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
25
|
Richards CJ, Ahmadi M, Stuart MCA, Kooi BJ, Åberg C, Roos WH. The effect of biomolecular corona on adsorption onto and desorption from a model lipid membrane. NANOSCALE 2022; 15:248-258. [PMID: 36472238 DOI: 10.1039/d2nr05296b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The current lack of insight into nanoparticle-cell membrane interactions hampers smart design strategies and thereby the development of effective nanodrugs. Quantitative and methodical approaches utilizing cell membrane models offer an opportunity to unravel particle-membrane interactions in a detailed manner under well controlled conditions. Here we use total internal reflection microscopy for real-time studies of the non-specific interactions between nanoparticles and a model cell membrane at 50 ms temporal resolution over a time course of several minutes. Maintaining a simple lipid bilayer system across conditions, adsorption and desorption were quantified as a function of biomolecular corona, particle size and fluid flow. The presence of a biomolecular corona reduced both the particle adsorption rate onto the membrane and the duration of adhesion, compared to pristine particle conditions. Particle size, on the other hand, was only observed to affect the adsorption rate. The introduction of flow reduced the number of adsorption events, but increased the residence time. Lastly, altering the composition of the membrane itself resulted in a decreased number of adsorption events onto negatively charged bilayers compared to neutral bilayers. Overall, a model membrane system offers a facile platform for real-time imaging of individual adsorption-desorption processes, revealing complex adsorption kinetics, governed by particle surface energy, size dependent interaction forces, flow and membrane composition.
Collapse
Affiliation(s)
- Ceri J Richards
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, 9747 AG Groningen, Netherlands.
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, Rijksuniversiteit Groningen, 9713 AV Groningen, Netherlands.
| | - Majid Ahmadi
- Nanostructure Materials and Interfaces, Zernike Institute for Advanced Materials, Rijksuniversiteit 9747 AG Groningen, Netherlands
| | - Marc C A Stuart
- Department of Electron Microscopy, Groningen Biomolecular Sciences and Biotechnology Institute, Rijksuniversiteit Groningen, 9747 AG Groningen, Netherlands
| | - Bart J Kooi
- Nanostructure Materials and Interfaces, Zernike Institute for Advanced Materials, Rijksuniversiteit 9747 AG Groningen, Netherlands
| | - Christoffer Åberg
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, Rijksuniversiteit Groningen, 9713 AV Groningen, Netherlands.
| | - Wouter H Roos
- Molecular Biophysics, Zernike Institute for Advanced Materials, Rijksuniversiteit Groningen, 9747 AG Groningen, Netherlands.
| |
Collapse
|
26
|
Münter R, Simonsen JB. Comment on "Optimal centrifugal isolating of liposome-protein complexes from human plasma" by L. Digiacomo, F. Giulimondi, A. L. Capriotti, S. Piovesana, C. M. Montone, R. Z. Chiozzi, A. Laganá, M. Mahmoudi, D. Pozzi and G. Caracciolo, Nanoscale Adv., 2021, 3, 3824. NANOSCALE ADVANCES 2022; 5:290-299. [PMID: 36605796 PMCID: PMC9765536 DOI: 10.1039/d2na00343k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/20/2022] [Indexed: 06/01/2023]
Abstract
In a recent paper in Nanoscale Advances, Digiacomo et al. conclude that centrifugation should be the method of choice for researchers who want to investigate the protein corona of liposomes for drug delivery in human plasma. In this Comment, we however propose the opposite - that centrifugation, in most cases, is unsuitable for isolating liposomes from human plasma. Our conclusion is based on the bulk literature on this and similar topics, and new experimental data based on formulations and protocols like the ones used by Digiacomo et al.
Collapse
Affiliation(s)
- Rasmus Münter
- Department of Health Technology, Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark (DTU) 2800 Kgs. Lyngby Denmark
| | - Jens B Simonsen
- Department of Health Technology, Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark (DTU) 2800 Kgs. Lyngby Denmark
| |
Collapse
|
27
|
Riedelová Z, de Los Santos Pereira A, Svoboda J, Pop-Georgievski O, Májek P, Pečánková K, Dyčka F, Rodriguez-Emmenegger C, Riedel T. The Relation Between Protein Adsorption and Hemocompatibility of Antifouling Polymer Brushes. Macromol Biosci 2022; 22:e2200247. [PMID: 35917216 DOI: 10.1002/mabi.202200247] [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: 06/15/2022] [Revised: 07/22/2022] [Indexed: 12/25/2022]
Abstract
Whenever an artificial surface comes into contact with blood, proteins are rapidly adsorbed onto its surface. This phenomenon, termed fouling, is then followed by a series of undesired reactions involving activation of complement or the coagulation cascade and adhesion of leukocytes and platelets leading to thrombus formation. Thus, considerable efforts are directed towards the preparation of fouling-resistant surfaces with the best possible hemocompatibility. Herein, a comprehensive hemocompatibility study after heparinized blood contact with seven polymer brushes prepared by surface-initiated atom transfer radical polymerization is reported. The resistance to fouling is quantified and thrombus formation and deposition of blood cellular components on the coatings are analyzed. Moreover, identification of the remaining adsorbed proteins is performed via mass spectroscopy to elucidate their influence on the surface hemocompatibility. Compared with an unmodified glass surface, the grafting of polymer brushes minimizes the adhesion of platelets and leukocytes and prevents the thrombus formation. The fouling from undiluted blood plasma is reduced by up to 99%. Most of the identified proteins are connected with the initial events of foreign body reaction towards biomaterial (coagulation cascade proteins, complement component, and inflammatory proteins). In addition, several proteins that are not previously linked with blood-biomaterial interaction are presented and discussed.
Collapse
Affiliation(s)
- Zuzana Riedelová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague, 162 06, Czech Republic
| | - Andres de Los Santos Pereira
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague, 162 06, Czech Republic
| | - Jan Svoboda
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague, 162 06, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague, 162 06, Czech Republic
| | - Pavel Májek
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague, 128 00, Czech Republic
| | - Klára Pečánková
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague, 128 00, Czech Republic
| | - Filip Dyčka
- Faculty of Science, University of South Bohemia, Branišovská 1760, České Budějovice, 370 05, Czech Republic
| | - Cesar Rodriguez-Emmenegger
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, Barcelona, 08028, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Spain.,DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, D-52074, Aachen, Germany
| | - Tomáš Riedel
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague, 162 06, Czech Republic
| |
Collapse
|
28
|
Palmieri V, Caracciolo G. Tuning the immune system by nanoparticle-biomolecular corona. NANOSCALE ADVANCES 2022; 4:3300-3308. [PMID: 36131704 PMCID: PMC9419885 DOI: 10.1039/d2na00290f] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/14/2022] [Indexed: 05/24/2023]
Abstract
Nanotechnology has a great potential to revolutionize the landscape of medicine, but an inadequate understanding of the nanomaterial-biological (nano-bio) interface hampers its ultimate clinical translation. Surface attachment of biomolecules provides a new biological identity of nanoparticles that plays a crucial role in vivo as it can activate the immune system triggering inflammatory responses, clearance from the body, and cellular toxicity. In this review, we summarize and critically analyze progress in understanding the relationship between the biological identity of nanoparticles and immune system activation. Accordingly, we discuss the implications of biomolecular corona on nanotoxicity, immune safety, and biocompatibility. We also highlight a perspective on engineering the biological identity of nanoparticles for modulating immunological responses.
Collapse
Affiliation(s)
- Valentina Palmieri
- Institute for Complex Systems, National Research Council of Italy Via dei Taurini 19 00185 Rome Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome Viale Regina Elena 291 00161 Rome Italy
| |
Collapse
|
29
|
Martin KE, Kalelkar PP, Coronel MM, Theriault HS, Schneider RS, García AJ. Host type 2 immune response to xenogeneic serum components impairs biomaterial-directed osteo-regenerative therapies. Biomaterials 2022; 286:121601. [PMID: 35660823 PMCID: PMC11458135 DOI: 10.1016/j.biomaterials.2022.121601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/21/2022]
Abstract
The transformative potential of cells as therapeutic agents is being realized in a wide range of applications, from regenerative medicine to cancer therapy to autoimmune disorders. The majority of these therapies require ex vivo expansion of the cellular product, often utilizing fetal bovine serum (FBS) in the culture media. However, the impact of residual FBS on immune responses to cell therapies and the resulting cell therapy outcomes remains unclear. Here, we show that hydrogel-delivered FBS elicits a robust type 2 immune response characterized by infiltration of eosinophils and CD4+ T cells. Host secretion of cytokines associated with type 2 immunity, including IL-4, IL-5, and IL-13, is also increased in FBS-containing hydrogels. We demonstrate that the immune response to xenogeneic serum components dominates the local environment and masks the immunomodulatory effects of biomaterial-delivered mesenchymal stromal/stem cells. Importantly, delivery of relatively small amounts of FBS (3.2% by volume) within BMP-2-containing biomaterial constructs dramatically reduces the ability of these constructs to promote de novo bone formation in a radial defect model in immunocompetent mice. These results urge caution when interpreting the immunological and tissue repair outcomes in immunocompetent pre-clinical models from cells and biomaterial constructs that have come in contact with xenogeneic serum components.
Collapse
Affiliation(s)
- Karen E Martin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav P Kalelkar
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - María M Coronel
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hannah S Theriault
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
30
|
Lee E, Lee M, Kwon S, Kim J, Kwon Y. Systematic and mechanistic analysis of AuNP-induced nanotoxicity for risk assessment of nanomedicine. NANO CONVERGENCE 2022; 9:27. [PMID: 35680772 PMCID: PMC9184696 DOI: 10.1186/s40580-022-00320-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/29/2022] [Indexed: 05/02/2023]
Abstract
For decades, nanoparticles (NPs) have been widely implemented in various biomedical fields due to their unique optical, thermal, and tunable properties. Particularly, gold nanoparticles (AuNPs) have opened new frontiers in sensing, targeted drug delivery, imaging, and photodynamic therapy, showing promising results for the treatment of various intractable diseases that affect quality of life and longevity. Despite the tremendous achievements of AuNPs-based approaches in biomedical applications, few AuNP-based nanomedicines have been evaluated in clinical trials, which is likely due to a shortage of understanding of the biological and pathological effects of AuNPs. The biological fate of AuNPs is tightly related to a variety of physicochemical parameters including size, shape, chemical structure of ligands, charge, and protein corona, and therefore evaluating the effects of these parameters on specific biological interactions is a major ongoing challenge. Therefore, this review focuses on ongoing nanotoxicology studies that aim to characterize the effect of various AuNP characteristics on AuNP-induced toxicity. Specifically, we focus on understanding how each parameter alters the specific biological interactions of AuNPs via mechanistic analysis of nano-bio interactions. We also discuss different cellular functions affected by AuNP treatment (e.g., cell motility, ROS generation, interaction with DNA, and immune response) to understand their potential human health risks. The information discussed herein could contribute to the safe usage of nanomedicine by providing a basis for appropriate risk assessment and for the development of nano-QSAR models.
Collapse
Affiliation(s)
- Euiyeon Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Minhyeong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - San Kwon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jongpil Kim
- Department of Chemistry, Dongguk University, Seoul, 04620, Korea.
| | - Youngeun Kwon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
31
|
Shaw J, Pearson RM. Nanoparticle personalized biomolecular corona: implications of pre-existing conditions for immunomodulation and cancer. Biomater Sci 2022; 10:2540-2549. [PMID: 35476072 PMCID: PMC9117514 DOI: 10.1039/d2bm00315e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Nanoparticles (NPs) have demonstrated great promise as immunotherapies for applications ranging from cancer, autoimmunity, and infectious disease. Upon encountering biological fluids, NPs rapidly adsorb biomolecules, forming the "biomolecular corona" (BC), and the altered character of NPs due to their newly acquired biological identity can impact their in vivo fate. Recently, it has been shown that the NP-BC is person-specific, and even minute differences in the biomolecule composition can give rise to altered immune recognition, cellular interactions, pharmacokinetics, and biodistribution. Given the current rise in the development of NP-based therapeutics, it is of utmost importance to better understand how pre-existing conditions, that result in the formation of a personalized BC, can be leveraged to aid in the prediction of the therapeutic outcomes of NPs. In this minireview, we will discuss the formation of the BC, implications of the BC for NP-biological interactions, and its clinical importance in the context of immunomodulation and cancer therapeutics.
Collapse
Affiliation(s)
- Jacob Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, USA.
| | - Ryan M Pearson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, USA.
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
Nienhaus K, Xue Y, Shang L, Nienhaus GU. Protein adsorption onto nanomaterials engineered for theranostic applications. NANOTECHNOLOGY 2022; 33:262001. [PMID: 35294940 DOI: 10.1088/1361-6528/ac5e6c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/15/2022] [Indexed: 06/14/2023]
Abstract
The key role of biomolecule adsorption onto engineered nanomaterials for therapeutic and diagnostic purposes has been well recognized by the nanobiotechnology community, and our mechanistic understanding of nano-bio interactions has greatly advanced over the past decades. Attention has recently shifted to gaining active control of nano-bio interactions, so as to enhance the efficacy of nanomaterials in biomedical applications. In this review, we summarize progress in this field and outline directions for future development. First, we briefly review fundamental knowledge about the intricate interactions between proteins and nanomaterials, as unraveled by a large number of mechanistic studies. Then, we give a systematic overview of the ways that protein-nanomaterial interactions have been exploited in biomedical applications, including the control of protein adsorption for enhancing the targeting efficiency of nanomedicines, the design of specific protein adsorption layers on the surfaces of nanomaterials for use as drug carriers, and the development of novel nanoparticle array-based sensors based on nano-bio interactions. We will focus on particularly relevant and recent examples within these areas. Finally, we conclude this topical review with an outlook on future developments in this fascinating research field.
Collapse
Affiliation(s)
- Karin Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), D-76131 Karlsruhe, Germany
| | - Yumeng Xue
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Li Shang
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Gerd Ulrich Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), D-76131 Karlsruhe, Germany
- Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), D-76344 Eggenstein-Leopoldshafen, Germany
- Institute of Biological and Chemical Systems, Karlsruhe Institute of Technology (KIT), D-76344 Eggenstein-Leopoldshafen, Germany
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| |
Collapse
|
33
|
Giulimondi F, Vulpis E, Digiacomo L, Giuli MV, Mancusi A, Capriotti AL, Laganà A, Cerrato A, Zenezini Chiozzi R, Nicoletti C, Amenitsch H, Cardarelli F, Masuelli L, Bei R, Screpanti I, Pozzi D, Zingoni A, Checquolo S, Caracciolo G. Opsonin-Deficient Nucleoproteic Corona Endows UnPEGylated Liposomes with Stealth Properties In Vivo. ACS NANO 2022; 16:2088-2100. [PMID: 35040637 PMCID: PMC8867903 DOI: 10.1021/acsnano.1c07687] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/14/2022] [Indexed: 05/21/2023]
Abstract
For several decades, surface grafted polyethylene glycol (PEG) has been a go-to strategy for preserving the synthetic identity of liposomes in physiological milieu and preventing clearance by immune cells. However, the limited clinical translation of PEGylated liposomes is mainly due to the protein corona formation and the subsequent modification of liposomes' synthetic identity, which affects their interactions with immune cells and blood residency. Here we exploit the electric charge of DNA to generate unPEGylated liposome/DNA complexes that, upon exposure to human plasma, gets covered with an opsonin-deficient protein corona. The final product of the synthetic process is a biomimetic nanoparticle type covered by a proteonucleotidic corona, or "proteoDNAsome", which maintains its synthetic identity in vivo and is able to slip past the immune system more efficiently than PEGylated liposomes. Accumulation of proteoDNAsomes in the spleen and the liver was lower than that of PEGylated systems. Our work highlights the importance of generating stable biomolecular coronas in the development of stealth unPEGylated particles, thus providing a connection between the biological behavior of particles in vivo and their synthetic identity.
Collapse
Affiliation(s)
- Francesca Giulimondi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Elisabetta Vulpis
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Luca Digiacomo
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Maria Valeria Giuli
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Angelica Mancusi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Anna Laura Capriotti
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Aldo Laganà
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Andrea Cerrato
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Riccardo Zenezini Chiozzi
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Carmine Nicoletti
- Unit
of Histology and Medical Embryology, Department of Anatomy, Histology,
Forensic Medicine and Orthopedics, Sapienza
University of Rome, Viale A. Scarpa 16, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute
of inorganic Chemistry, Graz University
of Technology, Stremayerg 6/IV, 8010 Graz, Austria
| | | | - Laura Masuelli
- Department
of Experimental Medicine, University of
Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberto Bei
- Department
of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Isabella Screpanti
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Daniela Pozzi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Alessandra Zingoni
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Saula Checquolo
- Department
of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy
| | - Giulio Caracciolo
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
34
|
Hu B, Liu R, Liu Q, Lin Z, Shi Y, Li J, Wang L, Li L, Xiao X, Wu Y. Engineering surface patterns on nanoparticles: New insights on nano-bio interactions. J Mater Chem B 2022; 10:2357-2383. [DOI: 10.1039/d1tb02549j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The surface properties of nanoparticles affect their fates in biological systems. Based on nanotechnology and methodology, pioneering works have explored the effects of chemical surface patterns on the behavior of...
Collapse
|
35
|
Marschall ALJ. Targeting the Inside of Cells with Biologicals: Chemicals as a Delivery Strategy. BioDrugs 2021; 35:643-671. [PMID: 34705260 PMCID: PMC8548996 DOI: 10.1007/s40259-021-00500-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Delivering macromolecules into the cytosol or nucleus is possible in vitro for DNA, RNA and proteins, but translation for clinical use has been limited. Therapeutic delivery of macromolecules into cells requires overcoming substantially higher barriers compared to the use of small molecule drugs or proteins in the extracellular space. Breakthroughs like DNA delivery for approved gene therapies and RNA delivery for silencing of genes (patisiran, ONPATTRO®, Alnylam Pharmaceuticals, Cambridge, MA, USA) or for vaccination such as the RNA-based coronavirus disease 2019 (COVID-19) vaccines demonstrated the feasibility of using macromolecules inside cells for therapy. Chemical carriers are part of the reason why these novel RNA-based therapeutics possess sufficient efficacy for their clinical application. A clear advantage of synthetic chemicals as carriers for macromolecule delivery is their favourable properties with respect to production and storage compared to more bioinspired vehicles like viral vectors or more complex drugs like cellular therapies. If biologicals can be applied to intracellular targets, the druggable space is substantially broadened by circumventing the limited utility of small molecules for blocking protein–protein interactions and the limitation of protein-based drugs to the extracellular space. An in depth understanding of the macromolecular cargo types, carrier types and the cell biology of delivery is crucial for optimal application and further development of biologicals inside cells. Basic mechanistic principles of the molecular and cell biological aspects of cytosolic/nuclear delivery of macromolecules, with particular consideration of protein delivery, are reviewed here. The efficiency of macromolecule delivery and applications in research and therapy are highlighted.
Collapse
Affiliation(s)
- Andrea L J Marschall
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Brunswick, Germany.
| |
Collapse
|
36
|
Onishchenko N, Tretiakova D, Vodovozova E. Spotlight on the protein corona of liposomes. Acta Biomater 2021; 134:57-78. [PMID: 34364016 DOI: 10.1016/j.actbio.2021.07.074] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/19/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Although an established drug delivery platform, liposomes have not fulfilled their true potential. In the body, interactions of liposomes are mediated by the layer of plasma proteins adsorbed on the surface, the protein corona. The review aims to collect the data of the last decade on liposome protein corona, tracing the path from interactions of individual proteins to the effects mediated by the protein corona in vivo. It offers a classification of the approaches to exploitation of the protein corona-rather than elimination thereof-based on the bilayer composition-corona composition-molecular interactions-biological performance framework. The multitude of factors that affect each level of this relationship urge to the widest implementation of bioinformatics tools to predict the most effective liposome compositions relying on the data on protein corona. Supplementing the picture with new pieces of accurately reported experimental data will contribute to the accuracy and efficiency of the predictions. STATEMENT OF SIGNIFICANCE: The review focuses on liposomes as an established nanomedicine platform and analyzes the available data on how the protein corona formed on liposome surface in biological fluids affects performance of the liposomes. The review offers a rigorous account of existing literature and critical analysis of methodology currently applied to the assessment of liposome-plasma protein interactions. It introduces a classification of the approaches to exploitation of the protein corona and tailoring liposome carriers to advance the field of nanoparticulate drug delivery systems for the benefit of patients.
Collapse
|
37
|
Voke E, Pinals RL, Goh NS, Landry MP. In Planta Nanosensors: Understanding Biocorona Formation for Functional Design. ACS Sens 2021; 6:2802-2814. [PMID: 34279907 PMCID: PMC10461777 DOI: 10.1021/acssensors.1c01159] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Climate change and population growth are straining agricultural output. To counter these changes and meet the growing demand for food and energy, the monitoring and engineering of crops are becoming increasingly necessary. Nanoparticle-based sensors have emerged in recent years as new tools to advance agricultural practices. As these nanoparticle-based sensors enter and travel through the complex biofluids within plants, biomolecules including proteins, metabolites, lipids, and carbohydrates adsorb onto the nanoparticle surfaces, forming a coating known as the "bio-corona". Understanding these nanoparticle-biomolecule interactions that govern nanosensor function in plants will be essential to successfully develop and translate nanoparticle-based sensors into broader agricultural practice.
Collapse
Affiliation(s)
- Elizabeth Voke
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
| | - Rebecca L Pinals
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
| | - Natalie S Goh
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
| | - Markita P Landry
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
- Innovative Genomics Institute (IGI), Berkeley, California 94720, United States
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, California 94720, United States
- Chan-Zuckerberg Biohub, San Francisco, California 94158, United States
| |
Collapse
|
38
|
Jang GJ, Jeong JY, Kang J, Cho W, Han SY. Size Dependence Unveiling the Adsorption Interaction of High-Density Lipoprotein Particles with PEGylated Gold Nanoparticles in Biomolecular Corona Formation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:9755-9763. [PMID: 34347501 DOI: 10.1021/acs.langmuir.1c01182] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Apolipoproteins have been often found to be highly enriched in the serum protein coronas produced on various engineered nanoparticles (NPs), which is also known to greatly influence the behaviors of protein corona NPs in the biological systems. As most of the apolipoproteins in blood are associated with lipoproteins, it suggests the active involvement of lipoproteins in the formation of biomolecular coronas on NPs. However, the interactions of lipoprotein complexes with NPs in the corona formation have been rarely understood. In this study, to obtain insights into the interactions, the formation of biomolecular coronas of high-density lipoproteins (HDLs) on the PEGylated gold NPs (PEG-AuNPs) of various sizes (20-150 nm dia.) was investigated as a model system. The results of this study revealed a noticeable size dependence, which is a drastic increase in the affinity of HDL for larger NPs and thus less-curved NP surfaces. For example, only a few HDLs per NP, which correspond to 5% surface coverage, were found to constitute the hard coronas of HDLs on 20 nm PEG-AuNPs, whereas 73% surface coverage was assessed for larger 150 nm PEG-AuNPs. However, the relative affinities of HDL and apolipoprotein A-1 (APOA1) examined in competition with human serum albumin exhibited the opposite size dependences, which suggests that the adsorption of HDLs is not driven by the constituent protein, APOA1. In fact, the total strength of non-covalent intermolecular interactions between a HDL particle and a NP relies on the physical contact between the two particles, which thus depends on the varying curvatures of spherical NPs in this case. Therefore, it was concluded that it is whole HDL complex that interacts with the spherical PEG-AuNPs in the initial stage of adsorption toward biomolecular coronas, which is unveiled by the distinct size dependence observed in this study.
Collapse
Affiliation(s)
- Gwi Ju Jang
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi 13120, Republic of Korea
| | - Ji Yeon Jeong
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi 13120, Republic of Korea
| | - Junghoon Kang
- Department of Chemistry, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Wonryeon Cho
- Department of Chemistry, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sang Yun Han
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi 13120, Republic of Korea
| |
Collapse
|
39
|
Yang M, Wu E, Tang W, Qian J, Zhan C. Interplay between nanomedicine and protein corona. J Mater Chem B 2021; 9:6713-6727. [PMID: 34328485 DOI: 10.1039/d1tb01063h] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanomedicine is recognized as a promising agent for diverse biomedical applications; however, its safety and efficiency in clinical practice remains to be enhanced. A priority issue is the protein corona (PC), which imparts unique biological identities to prototype and determines the actual biological functions in biological fluids. Decades of work has already illuminated abundant considerations that influence the composition of the protein corona. Thereinto, the physical assets of nanomedicines (e.g., size and shape, surface properties, nanomaterials) and the biological environment collectively play fundamental roles in shaping the PC, including the types and quantities of plasma proteins. The properties of nanomedicines are dependent on certain factors. This review aims to explore the applications of nanomedicines by regulating their interplay with PC.
Collapse
Affiliation(s)
- Min Yang
- Department of Pharmacology, School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China.
| | - Ercan Wu
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Wenjing Tang
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Jun Qian
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China. and MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
40
|
Digiacomo L, Giulimondi F, Capriotti AL, Piovesana S, Montone CM, Chiozzi RZ, Laganà A, Mahmoudi M, Pozzi D, Caracciolo G. Optimal centrifugal isolating of liposome-protein complexes from human plasma. NANOSCALE ADVANCES 2021; 3:3824-3834. [PMID: 36133013 PMCID: PMC9418580 DOI: 10.1039/d1na00211b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/15/2021] [Indexed: 05/14/2023]
Abstract
In the past few years, characterization of the protein corona (PC) that forms around liposomal systems has gained increasing interest for the development of novel therapeutic and diagnostic technologies. At the crossroads of fast-moving research fields, the interdisciplinarity of protein corona investigations poses challenges for experimental design and reporting. Isolation of liposome-protein complexes from biological fluids has been identified as a fundamental step of the entire workflow of PC characterization but exact specifications for conditions to optimize pelleting remain elusive. In the present work, key factors affecting precipitation of liposome-protein complexes by centrifugation, including time of centrifugation, total sample volume, lipid : protein ratio and contamination from biological NPs were comprehensively evaluated. Here we show that the total amount of isolated liposome-protein complexes and the extent of contamination from biological NPs may vary with influence factors. Our results provide protein corona researchers with precise indications to separate liposome-protein complexes from protein-rich fluids and include proper controls, thus they are anticipated to catalyze improved consistency of data mining and computational modelling of protein corona composition.
Collapse
Affiliation(s)
- Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome Viale Regina Elena 291 00161 Rome Italy
| | - Francesca Giulimondi
- Department of Molecular Medicine, Sapienza University of Rome Viale Regina Elena 291 00161 Rome Italy
| | - Anna Laura Capriotti
- Department of Chemistry, Sapienza University of Rome P.le A. Moro 5 00185 Rome Italy
| | - Susy Piovesana
- Department of Chemistry, Sapienza University of Rome P.le A. Moro 5 00185 Rome Italy
| | - Carmela Maria Montone
- Department of Chemistry, Sapienza University of Rome P.le A. Moro 5 00185 Rome Italy
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences Utrecht University Heidelberglaan 8 3584 CS Utrecht The Netherlands
| | - Aldo Laganà
- Department of Chemistry, Sapienza University of Rome P.le A. Moro 5 00185 Rome Italy
| | - Morteza Mahmoudi
- Department of Radiology, Precision Health Program, Michigan State University MI USA
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome Viale Regina Elena 291 00161 Rome Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome Viale Regina Elena 291 00161 Rome Italy
| |
Collapse
|
41
|
A Proteomic Study on the Personalized Protein Corona of Liposomes. Relevance for Early Diagnosis of Pancreatic DUCTAL Adenocarcinoma and Biomarker Detection. JOURNAL OF NANOTHERANOSTICS 2021. [DOI: 10.3390/jnt2020006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Due to late diagnosis, high incidence of metastasis, and poor survival rate, pancreatic cancer is one of the most leading cause of cancer-related death. Although manifold recent efforts have been done to achieve an early diagnosis of pancreatic cancer, CA-19.9 is currently the unique biomarker that is adopted for the detection, despite its limits in terms of sensitivity and specificity. To identify potential protein biomarkers for pancreatic ductal adenocarcinoma (PDAC), we used three model liposomes as nanoplatforms that accumulate proteins from human plasma and studied the composition of this biomolecular layer, which is known as protein corona. Indeed, plasma proteins adsorb on nanoparticle surface according to their abundance and affinity to the employed nanomaterial, thus even small differences between healthy and PDAC protein expression levels can be, in principle, detected. By mass spectrometry experiments, we quantified such differences and identified possible biomarkers for PDAC. Some of them are already known to exhibit different expressions in PDAC proteomes, whereas the role of other relevant proteins is still not clear. Therefore, we predict that the employment of nanomaterials and their protein corona may represent a useful tool to amplify the detection sensitivity of cancer biomarkers, which may be used for the early diagnosis of PDAC, with clinical implication for the subsequent therapy in the context of personalized medicine.
Collapse
|
42
|
Liessi N, Maragliano L, Castagnola V, Bramini M, Benfenati F, Armirotti A. Isobaric Labeling Proteomics Allows a High-Throughput Investigation of Protein Corona Orientation. Anal Chem 2020; 93:784-791. [PMID: 33285070 PMCID: PMC7818227 DOI: 10.1021/acs.analchem.0c03134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
formation of the biomolecular corona represents a crucial factor
in controlling the biological interactions and trafficking of nanomaterials.
In this context, the availability of key epitopes exposed on the surface
of the corona, and able to engage the biological machinery, is important
to define the biological fate of the material. While the full biomolecular
corona composition can be investigated by conventional bottom-up proteomics,
the assessment of the spatial orientation of proteins in the corona
in a high-throughput fashion is still challenging. In this work, we
show that labeling corona proteins with isobaric tags in their native
conditions and analyzing the MS/MS spectra of tryptic peptides allow
an easy and high-throughput assessment of the inner/outer orientation
of the corresponding proteins in the original corona. We put our results
in the context of what is currently known of the protein corona of
graphene-based nanomaterials. Our conclusions are in line with previous
data and were confirmed by in silico calculations.
Collapse
Affiliation(s)
- Nara Liessi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,Department of Applied Physics, Faculty of Sciences, University of Granada, Fuente Nueva s/n, 18071 Granada, Spain
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
43
|
Liu C, Zhang L, Zhu W, Guo R, Sun H, Chen X, Deng N. Barriers and Strategies of Cationic Liposomes for Cancer Gene Therapy. Mol Ther Methods Clin Dev 2020; 18:751-764. [PMID: 32913882 PMCID: PMC7452052 DOI: 10.1016/j.omtm.2020.07.015] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cationic liposomes (CLs) have been regarded as the most promising gene delivery vectors for decades with the advantages of excellent biodegradability, biocompatibility, and high nucleic acid encapsulation efficiency. However, the clinical use of CLs in cancer gene therapy is limited because of many uncertain factors in vivo. Extracellular barriers such as opsonization, rapid clearance by the reticuloendothelial system and poor tumor penetration, and intracellular barriers, including endosomal/lysosomal entrapped network and restricted diffusion to the nucleus, make CLs not the ideal vector for transferring extrinsic genes in the body. However, the obstacles in achieving productive therapeutic effects of nucleic acids can be addressed by tailoring the properties of CLs, which are influenced by lipid compositions and surface modification. This review focuses on the physiological barriers of CLs against cancer gene therapy and the effects of lipid compositions on governing transfection efficiency, and it briefly discusses the impacts of particle size, membrane charge density, and surface modification on the fate of CLs in vivo, which may provide guidance for their preclinical studies.
Collapse
Affiliation(s)
- Chunyan Liu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Ligang Zhang
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Wenhui Zhu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Raoqing Guo
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Huamin Sun
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Xi Chen
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| | - Ning Deng
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
44
|
Sharifi S, Caracciolo G, Mahmoudi M. Biomolecular Corona Affects Controlled Release of Drug Payloads from Nanocarriers. Trends Pharmacol Sci 2020; 41:641-652. [DOI: 10.1016/j.tips.2020.06.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
|
45
|
Simonsen JB, Münter R. Pay Attention to Biological Nanoparticles when Studying the Protein Corona on Nanomedicines. Angew Chem Int Ed Engl 2020; 59:12584-12588. [DOI: 10.1002/anie.202004611] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Jens B. Simonsen
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| | - Rasmus Münter
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| |
Collapse
|
46
|
Simonsen JB, Münter R. Pay Attention to Biological Nanoparticles when Studying the Protein Corona on Nanomedicines. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jens B. Simonsen
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| | - Rasmus Münter
- Department of Health Technology Technical University of Denmark 2800 Kgs. Lyngby Denmark
| |
Collapse
|
47
|
Immunoglobulins on the surface of differently charged polymer nanoparticles. Biointerphases 2020; 15:031009. [DOI: 10.1116/6.0000139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
48
|
Liu N, Tang M, Ding J. The interaction between nanoparticles-protein corona complex and cells and its toxic effect on cells. CHEMOSPHERE 2020; 245:125624. [PMID: 31864050 DOI: 10.1016/j.chemosphere.2019.125624] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 05/23/2023]
Abstract
Once nanoparticles (NPs) contact with the biological fluids, the proteins immediately adsorb onto their surface, forming a layer called protein corona (PC), which bestows the biological identity on NPs. Importantly, the NPs-PC complex is the true identity of NPs in physiological environment. Based on the affinity and the binding and dissociation rate, PC is classified into soft protein corona, hard protein corona, and interfacial protein corona. Especially, the hard PC, a protein layer relatively stable and closer to their surface, plays particularly important role in the biological effects of the complex. However, the abundant corona proteins rarely correspond to the most abundant proteins found in biological fluids. The composition profile, formation and conformational change of PC can be affected by many factors. Here, the influence factors, not only the nature of NPs, but also surface chemistry and biological medium, are discussed. Likewise, the formed PC influences the interaction between NPs and cells, and the associated subsequent cellular uptake and cytotoxicity. The uncontrolled PC formation may induce undesirable and sometimes opposite results: increasing or inhibiting cellular uptake, hindering active targeting or contributing to passive targeting, mitigating or aggravating cytotoxicity, and stimulating or mitigating the immune response. In the present review, we discuss these aspects and hope to provide a valuable reference for controlling protein adsorption, predicting their behavior in vivo experiments and designing lower toxicity and enhanced targeting nanomedical materials for nanomedicine.
Collapse
Affiliation(s)
- Na Liu
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| | - Meng Tang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| | - Jiandong Ding
- Department of Cardiology, Zhongda Hospital, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| |
Collapse
|
49
|
Yang K, Mesquita B, Horvatovich P, Salvati A. Tuning liposome composition to modulate corona formation in human serum and cellular uptake. Acta Biomater 2020; 106:314-327. [PMID: 32081780 DOI: 10.1016/j.actbio.2020.02.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
Nano-sized objects such as liposomes are modified by adsorption of biomolecules in biological fluids. The resulting corona critically changes nanoparticle behavior at cellular level. A better control of corona composition could allow to modulate uptake by cells. Within this context, in this work, liposomes of different charge were prepared by mixing negatively charged and zwitterionic lipids to different ratios. The series obtained was used as a model system with tailored surface properties to modulate corona composition and determine the effects on liposome interactions with cells. Uptake efficiency and uptake kinetics of the different liposomes were determined by flow cytometry and fluorescence imaging. Particular care was taken in optimizing the methods to isolate the corona forming in human serum to prevent liposome agglomeration and to exclude residual free proteins, which could confuse the results. Thanks to the optimized methods, mass spectrometry of replicate corona isolations showed excellent reproducibility and this allowed semi-quantitative analysis to determine for each formulation the most abundant proteins in the corona. The results showed that by changing the fraction of zwitterionic and charged lipids in the bilayer, the amount and identity of the most abundant proteins adsorbed from serum differed. Interestingly, the formulations also showed very different uptake kinetics. Similar approaches can be used to tune lipid composition in a systematic way in order to obtain formulations with the desired corona and cell uptake behavior. STATEMENT OF SIGNIFICANCE: Liposomes and other nano-sized objects when introduced in biological fluids are known to adsorb biomolecules forming the so-called nanoparticle corona. This layer strongly affects the subsequent interactions of liposomes with cells. Here, by tuning lipid composition in a systematic way, a series of liposomes with tailored surface properties has been prepared to modulate the corona forming in human serum. Liposomes with very different cellular uptake kinetics have been obtained and their corona was identified in order to determine the most enriched proteins on the different formulations. By combining corona composition and uptake kinetics candidate corona proteins associated with reduced or increased uptake by cells can be identified and the liposome formulation can be tuned to obtain the desired uptake behavior.
Collapse
Affiliation(s)
- Keni Yang
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Bárbara Mesquita
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, the Netherlands
| | - Anna Salvati
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, the Netherlands.
| |
Collapse
|
50
|
Quagliarini E, Di Santo R, Palchetti S, Ferri G, Cardarelli F, Pozzi D, Caracciolo G. Effect of Protein Corona on The Transfection Efficiency of Lipid-Coated Graphene Oxide-Based Cell Transfection Reagents. Pharmaceutics 2020; 12:E113. [PMID: 32019150 PMCID: PMC7076454 DOI: 10.3390/pharmaceutics12020113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/13/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Coating graphene oxide nanoflakes with cationic lipids leads to highly homogeneous nanoparticles (GOCL NPs) with optimised physicochemical properties for gene delivery applications. In view of in vivo applications, here we use dynamic light scattering, micro-electrophoresis and one-dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis to explore the bionano interactions between GOCL/DNA complexes (hereafter referred to as "grapholipoplexes") and human plasma. When exposed to increasing protein concentrations, grapholipoplexes get covered by a protein corona that evolves with protein concentration, leading to biocoronated complexes with modified physicochemical properties. Here, we show that the formation of a protein corona dramatically changes the interactions of grapholipoplexes with four cancer cell lines: two breast cancer cell lines (MDA-MB and MCF-7 cells), a malignant glioma cell line (U-87 MG) and an epithelial colorectal adenocarcinoma cell line (CACO-2). Luciferase assay clearly indicates a monotonous reduction of the transfection efficiency of biocoronated grapholipoplexes as a function of protein concentration. Finally, we report evidence that a protein corona formed at high protein concentrations (as those present in in vivo studies) promotes a higher capture of biocoronated grapholipoplexes within degradative intracellular compartments (e.g., lysosomes), with respect to their pristine counterparts. On the other hand, coronas formed at low protein concentrations (human plasma = 2.5%) lead to high transfection efficiency with no appreciable cytotoxicity. We conclude with a critical assessment of relevant perspectives for the development of novel biocoronated gene delivery systems.
Collapse
Affiliation(s)
- Erica Quagliarini
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy;
| | - Riccardo Di Santo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (R.D.S.); (S.P.)
| | - Sara Palchetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (R.D.S.); (S.P.)
| | - Gianmarco Ferri
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (F.C.)
| | - Francesco Cardarelli
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (F.C.)
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (R.D.S.); (S.P.)
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy; (R.D.S.); (S.P.)
| |
Collapse
|