1
|
Li Z, Xiao C, Yang X, Li Z. Progress in the mechanical properties of nanoparticles for tumor-targeting delivery. Chem Soc Rev 2025. [PMID: 40341776 DOI: 10.1039/d3cs00912b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Cancer nanomedicines have attracted significant attention in the past several decades, and the physicochemical properties, such as the size, shape, composition, surface charge, hydrophobicity, and mechanical properties, of nanoparticles have been optimized for potent cancer therapy. Since publishing our 2020 tutorial review "Influence of nanomedicine mechanical properties on tumor targeting delivery" in Chemical Society Reviews, substantial advancements have been made in understanding the role of mechanical properties in cancer nanomedicine. Notably, in vivo transport processes that are dependent on the mechanical properties of nanomedicine, including long circulation, tumor accumulation, and deep penetration, have been extensively studied using various nano-drug delivery systems. These studies have demonstrated that leveraging these mechanical properties can significantly enhance the antitumor efficacy of nanomedicine. In this review, we categorize the advancements in the mechanical properties of cancer nanomedicine into three distinct themes: the interactions between nanoparticles with varied mechanical properties and cells (2002 - present), the impact of these properties on in vivo delivery processes (2007 - present), and the strategic use of mechanical properties to boost cancer therapy (2023 - present). We analyze how different mechanical properties of organic, inorganic, hybrid, and biological nanoparticles affect their delivery processes at the macroscopic level, i.e., in tissues, organs and cells. At the microscopic level, their biological and physical interactions with biological barriers, physiological structures, cell membranes, organelles, and other structures reveal the potential mechanism of nanoparticles' mechanical properties in determining their antitumor efficacy. Furthermore, we address the current challenges and future prospects in the mechanical properties of cancer nanomedicine, as well as the clinical translation potential of nanoparticles with diverse mechanical characteristics.
Collapse
Affiliation(s)
- Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
2
|
Lu C, Li C, Gu N, Yang F. Emerging Elastic Micro-Nano Materials for Diagnosis and Treatment of Thrombosis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0614. [PMID: 40028043 PMCID: PMC11868703 DOI: 10.34133/research.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Thrombus is a blood clot that forms in a blood vessel at the point of flaking. Thrombosis is closely associated with cardiovascular diseases caused by different sources and factors. However, the current clinical methods of thrombus diagnosis and treatment still have problems with targeting, permeability, stability, and biosafety. Therefore, in recent years, based on the development of micro/nano technology, researchers have tried to develop some new strategies for the diagnosis and treatment of thrombosis. Due to the unique structural characteristics, the micro-nano materials in physiological environments show excellent transport and delivery properties such as better in vivo circulation, longer life span, better targeting ability, and controllable cellular internalization. Especially, elasticity and stiffness are inherent mechanical properties of some well-designed micro-nano materials, which can make them better adapted to the needs of thrombosis diagnosis and treatment. Herein, this review first introduces the thrombotic microenvironment to characterize the thrombus development process. Then, to fine-tune the pathological occurrence and development of thrombosis, the role of elastic micro-nano materials for thrombus diagnosis and treatment is summarized. The properties, preparation methods, and biological fate of these materials have been discussed in detail. Following, the applications of elastic micro-nano materials in biomedical imaging, drug delivery, and therapy of thrombosis are highlighted. Last, the shortcomings and future design strategies of elastic micro-nano materials in diagnosis and treatment of clinical thrombosis are discussed. This review will provide new ideas for the use of nanotechnology in clinical diagnosis and treatment of thrombus in the future.
Collapse
Affiliation(s)
- Chenxin Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| | - Chunjian Li
- Department of Cardiology,
The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School,
Nanjing University, Nanjing 210093, P. R. China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
3
|
Lee JK, Guevara V, Akanbi OD, Hoff JD, Kupor D, Brannon ER, Eniola-Adefeso O. Deciphering neutrophil dynamics: Enhanced phagocytosis of elastic particles and impact on vascular-targeted carrier performance. SCIENCE ADVANCES 2025; 11:eadp1461. [PMID: 39752488 PMCID: PMC11698085 DOI: 10.1126/sciadv.adp1461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025]
Abstract
Particle elasticity has widely been established to substantially influence immune cell clearance and circulation time of vascular-targeted carriers (VTCs). However, prior studies have primarily investigated interactions with macrophages, monocytic cell lines, and in vivo murine models. Interactions between particles and human neutrophils remain largely unexplored, although they represent a critical aspect of VTC performance. Here, we explore the impact of particle elasticity on primary human neutrophil phagocytosis using polyethylene glycol-based particles of different elastic moduli. We found that neutrophils effectively phagocytose deformable particles irrespective of their modulus, indicating a departure from established phagocytosis trends seen with other types of immune cells. These findings highlight the observed phenotypic difference between different types of phagocytes and underscore the need to characterize VTC performance using various cell types and animal models that represent human systems closely.
Collapse
Affiliation(s)
- Jonathan K. Lee
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Valentina Guevara
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Oluwaseun D. Akanbi
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - J. Damon Hoff
- Small Molecule Analysis Group, Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Kupor
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
4
|
Fang JY, Yang Z, Hu W, Hoang BX, Han B. Viscoelastic Hydrogel Modulates Phenotype of Macrophage-Derived Multinucleated Cells and Macrophage Differentiation in Foreign Body Reactions. J Biomed Mater Res A 2025; 113:e37814. [PMID: 39429027 DOI: 10.1002/jbm.a.37814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/23/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Biomaterial-induced macrophage-derived multinucleated cells (MNCs) are often observed at or near material implantation sites, yet their subtypes and roles in tissue repair and wound healing remain unclear. This study compares material-induced MNCs to cytokine-induced MNCs using both in vitro and in vivo models. 3D-embedded Raw264.7 cells and rat bone marrow-derived monocytes (BMDMs), with or without cytokines such as IL-4 and RANKL, were characterized for their MNC morphologies and subtypes via in situ immunocytochemistry and flow cytometry. Macrophage polarization and osteoclastic differentiation were assessed through NO production, arginase activity, and tartrate-resistant acid phosphatase levels. 3D matrix-induced MNCs expressed the same phenotypic heterogeneity as the IL-4 and RANK-treated ones. 3D matrix-induced MNCs displayed the same phenotypic heterogeneity as those treated with IL-4 and RANKL. A high viscoelastic matrix (1006.48 ± 92.29 Pa) induced larger populations of proinflammatory and osteoclast-like MNCs, whereas a low viscoelastic matrix (38.61 ± 7.56 Pa) supported active differentiation and gene expression across pro-, anti-inflammatory, and osteoclast-like macrophages. Matrix viscoelasticity also influenced the effects of IL-4 and RANKL on macrophage-derived MNC polarization. In an in vivo subcutaneous implantation model, medium to high viscoelastic matrices exhibited higher populations of CD86+ and RANK+ MNCs, while low viscoelastic matrices showed higher populations of CD206+ MNCs. These findings suggest that matrix viscoelasticity modulates macrophage differentiation and MNC phenotype, with low viscoelastic matrices potentially favoring anti-inflammatory MNCs and macrophage differentiation suitable for subcutaneous implantation.
Collapse
Affiliation(s)
- Josephine Y Fang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Craniofacial Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, USA
| | - Zhi Yang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Wei Hu
- M.C. Gill Composites Center, University of Southern California, Los Angeles, USA
| | - Ba Xuan Hoang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Bo Han
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Biomedical Enginnering, Viterbi School of Engineering, University of Southern California, Los Angeles, USA
| |
Collapse
|
5
|
Ando M, Horonushi D, Yuki H, Kato S, Yoshida A, Yasuda K. Spatial Discrimination Limit Analysis of Macrophage Phagocytosis Between Target Antigens and Non-Target Objects Using Microcapillary Manipulation Assay. MICROMACHINES 2024; 15:1394. [PMID: 39597206 PMCID: PMC11596049 DOI: 10.3390/mi15111394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
During phagocytosis, the FcGR-IgG bond is thought to be necessary to promote cell-membrane extension as the zipper mechanism. However, does this zipper mechanism provide a spatial antigen discrimination capability that allows macrophages to selectively phagocytose only antigens, especially for clusters with a mixture of antigens and non-antigens? To elucidate the ability and limitation of the zipper mechanism, we fed a coupled 2 μm IgG-coated and 4.5 μm non-coated polystyrene bead mixtures to macrophages and observed their phagocytosis. Macrophage engulfed the mixed clusters, including the 4.5 μm non-coated polystyrene part, indicating that the non-coated particles can be engulfed even without the zipper mechanism as far as coupled to the opsonized particles. In contrast, when the non-opsonized particle part was held by the microcapillary manipulation assay, macrophages pinched off the non-coated polystyrene particle part and internalized the opsonized particle part only. The results suggest that (1) an IgG-coated surface is needed to anchor phagocytosis by cell-membrane protrusion; however, (2) once the antibody-dependent cell phagocytosis is started, phagocytosis can proceed with the uncoated objects as the followers of the internalizing opsonized particles even without the support of the zipper mechanism. They may also indicate the concern of misleading the immune system to target unexpected objects because of their aggregation with target pathogens and the possibility of new medical applications to capture the non-opsonized target objects by the aggregation with small antigens to activate an immune response.
Collapse
Affiliation(s)
- Maiha Ando
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; (M.A.); (D.H.); (H.Y.); (A.Y.)
| | - Dan Horonushi
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; (M.A.); (D.H.); (H.Y.); (A.Y.)
| | - Haruka Yuki
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; (M.A.); (D.H.); (H.Y.); (A.Y.)
| | - Shinya Kato
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan;
| | - Amane Yoshida
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; (M.A.); (D.H.); (H.Y.); (A.Y.)
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; (M.A.); (D.H.); (H.Y.); (A.Y.)
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan;
| |
Collapse
|
6
|
Settle AH, Winer BY, de Jesus MM, Seeman L, Wang Z, Chan E, Romin Y, Li Z, Miele MM, Hendrickson RC, Vorselen D, Perry JSA, Huse M. β2 integrins impose a mechanical checkpoint on macrophage phagocytosis. Nat Commun 2024; 15:8182. [PMID: 39294148 PMCID: PMC11411054 DOI: 10.1038/s41467-024-52453-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
Phagocytosis is an intensely physical process that depends on the mechanical properties of both the phagocytic cell and its chosen target. Here, we employed differentially deformable hydrogel microparticles to examine the role of cargo rigidity in the regulation of phagocytosis by macrophages. Whereas stiff cargos elicited canonical phagocytic cup formation and rapid engulfment, soft cargos induced an architecturally distinct response, characterized by filamentous actin protrusions at the center of the contact site, slower cup advancement, and frequent phagocytic stalling. Using phosphoproteomics, we identified β2 integrins as critical mediators of this mechanically regulated phagocytic switch. Macrophages lacking β2 integrins or their downstream effectors, Talin1 and Vinculin, exhibited specific defects in phagocytic cup architecture and selective suppression of stiff cargo uptake. We conclude that integrin signaling serves as a mechanical checkpoint during phagocytosis to pair cargo rigidity to the appropriate mode of engulfment.
Collapse
Affiliation(s)
- Alexander H Settle
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Y Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel M de Jesus
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauren Seeman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaoquan Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Eric Chan
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoning Li
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL, USA
| | - Daan Vorselen
- Cell Biology and Immunology, Wageningen University & Research, Wageningen, The Netherlands
| | - Justin S A Perry
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Morgan Huse
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
7
|
Oguma T, Kanazawa T, Kaneko YK, Sato R, Serizawa M, Ooka A, Yamaguchi M, Ishikawa T, Kondo H. Effects of phospholipid type and particle size on lipid nanoparticle distribution in vivo and in pancreatic islets. J Control Release 2024; 373:917-928. [PMID: 39079658 DOI: 10.1016/j.jconrel.2024.07.059] [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: 03/15/2024] [Revised: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024]
Abstract
Lipid nanoparticles (LNPs) have recently been used as nanocarriers in drug delivery systems for nucleic acid drugs. Their practical applications are currently primarily limited to the liver and specific organs. However, altering the type and composition ratio of phospholipids improves their distribution in organs other than the liver, such as the spleen and lungs. This study aimed to elucidate the effects of LNP components and particle size on in vivo distribution through systemic circulation to pancreatic islets to achieve better targeting of islets, which are a fundamental therapeutic target for diabetes. Fluorescence-labeled LNPs were prepared using three phospholipids: 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), with particle sizes of 30-160 nm (diameter) using a microfluidic device. Baffled-structured iLiNP devices with adjusted flow-rate ratios and total flow rates were used. After the intravenous administration of LNPs to C57BL/6 J mice, the distribution of each LNP type to the major organs, including the pancreas and pancreatic islets, was compared using ex vivo fluorescence imaging and observation of pancreatic tissue sections. DSPC-LNPs- and DOPE-LNPs showed the highest distribution in the spleen and liver, respectively. In contrast, the DOPC-LNPs showed the highest distribution in the pancreas and the lowest distribution in the liver and spleen. In addition, smaller particles showed better distribution throughout the pancreas. The most significant LNP distribution in the islets was observed for DOPC-LNPs with a particle size of 160 nm. Furthermore, larger LNPs tended to be distributed in the islets, whereas smaller LNPs tended to be distributed in the exocrine glands. DOPC-LNPs were distributed in the islets at all cholesterol concentrations, with a high distribution observed at >40% cholesterol and > 3% PEG and the distribution was higher at 24 h than at 4 h. Thus, LNP composition and particle size significantly affected islet distribution characteristics, indicating that DOPC-LNPs may be a drug delivery system for effectively targeting the pancreas and islets.
Collapse
Affiliation(s)
- Takayuki Oguma
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Center for Clinical Research, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takanori Kanazawa
- Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Department of Clinical Pharmacology, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Shoumachi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Shoumachi, Tokushima 770-8505, Japan.
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Ren Sato
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Miku Serizawa
- Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Akira Ooka
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiromu Kondo
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
8
|
Wang X, Hao X, Zhang Y, Wu Q, Zhou J, Cheng Z, Chen J, Liu S, Pan J, Wang Y, Fan JB. Bioinspired Adaptive Microdrugs Enhance the Chemotherapy of Malignant Glioma: Beyond Their Nanodrugs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405165. [PMID: 38758975 DOI: 10.1002/adma.202405165] [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: 04/10/2024] [Revised: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Solid nanoparticle-mediated drug delivery systems are usually confined to nanoscale due to the enhanced permeability and retention effect. However, they remain a great challenge for malignant glioma chemotherapy because of poor drug delivery efficiency and insufficient tumor penetration resulting from the blood-brain barrier/blood-brain tumor barrier (BBB/BBTB). Inspired by biological microparticles (e.g., cells) with excellent adaptive deformation, it is demonstrated that the adaptive microdrugs (even up to 3.0 µm in size) are more efficient than their nanodrugs (less than 200 nm in size) to cross BBB/BBTB and penetrate into tumor tissues, achieving highly efficient chemotherapy of malignant glioma. The distinct delivery of the adaptive microdrugs is mainly attributed to the enhanced interfacial binding and endocytosis via adaptive deformation. As expected, the obtained adaptive microdrugs exhibit enhanced accumulation, deep penetration, and cellular internalization into tumor tissues in comparison with nanodrugs, significantly improving the survival rate of glioblastoma mice. It is believed that the bioinspired adaptive microdrugs enable them to efficiently cross physiological barriers and deeply penetrate tumor tissues for drug delivery, providing an avenue for the treatment of solid tumors.
Collapse
Affiliation(s)
- Xuejiao Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xiangrong Hao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yangning Zhang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qun Wu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jiajia Zhou
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510515, P. R. China
| | - Zhongman Cheng
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jianping Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
- Department of Radiotherapy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, P. R. China
| | - Sijia Liu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jiahao Pan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Ying Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jun-Bing Fan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
9
|
Mei F, Guo Y, Wang Y, Zhou Y, Heng BC, Xie M, Huang X, Zhang S, Ding S, Liu F, Deng X, Chen L, Yang C. Matrix stiffness regulates macrophage polarisation via the Piezo1-YAP signalling axis. Cell Prolif 2024; 57:e13640. [PMID: 38556840 PMCID: PMC11294424 DOI: 10.1111/cpr.13640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
Macrophages play a pivotal role in the immunological cascade activated in response to biomedical implants, which predetermine acceptance or rejection of implants by the host via pro- and anti-inflammatory polarisation states. The role of chemical signals in macrophage polarisation is well-established, but how physical cues regulate macrophage function that may play a fundamental role in implant-bone interface, remains poorly understood. Here we find that bone marrow-derived macrophages (BMDM) cultured on polyacrylamide gels of varying stiffness exhibit different polarisation states. BMDM are 'primed' to a pro-inflammatory M1 phenotype on stiff substrates, while to an anti-inflammatory M2 phenotype on soft and medium stiffness substrates. It is further observed that matrix stiffening increases Piezo1 expression, as well as leads to subsequent activation of the mechanotransduction signalling effector YAP, thus favouring M1 polarisation whilst suppressing M2 polarisation. Moreover, upon treatment with YAP inhibitor, we successfully induce macrophage re-polarisation to the M2 state within the implant site microenvironment, which in turn promotes implant osseointegration. Collectively, our present study thus characterises the critical role of the Piezo1-YAP signalling axis in macrophage mechanosensing and stiffness-mediated macrophage polarisation and provides cues for the design of immuno-modulatory biomaterials that can regulate the macrophage phenotype.
Collapse
Affiliation(s)
- Feng Mei
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Yaru Guo
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingChina
| | - Yu Wang
- Department of OrthodonticsPeking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital StomatologyBeijingChina
| | - Yingying Zhou
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijingChina
| | - Boon Chin Heng
- Central LaboratoryPeking University School and Hospital of StomatologyBeijingChina
| | - Mengru Xie
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Xiaofei Huang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Shihan Zhang
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingChina
| | - Shuai Ding
- Department of OrthodonticsPeking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital StomatologyBeijingChina
| | - Fangyong Liu
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijingChina
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingChina
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijingChina
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Cheng Yang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
10
|
ten Brink T, Damanik F, Rotmans JI, Moroni L. Unraveling and Harnessing the Immune Response at the Cell-Biomaterial Interface for Tissue Engineering Purposes. Adv Healthc Mater 2024; 13:e2301939. [PMID: 38217464 PMCID: PMC11468937 DOI: 10.1002/adhm.202301939] [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: 06/19/2023] [Revised: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Biomaterials are defined as "engineered materials" and include a range of natural and synthetic products, designed for their introduction into and interaction with living tissues. Biomaterials are considered prominent tools in regenerative medicine that support the restoration of tissue defects and retain physiologic functionality. Although commonly used in the medical field, these constructs are inherently foreign toward the host and induce an immune response at the material-tissue interface, defined as the foreign body response (FBR). A strong connection between the foreign body response and tissue regeneration is suggested, in which an appropriate amount of immune response and macrophage polarization is necessary to trigger autologous tissue formation. Recent developments in this field have led to the characterization of immunomodulatory traits that optimizes bioactivity, the integration of biomaterials and determines the fate of tissue regeneration. This review addresses a variety of aspects that are involved in steering the inflammatory response, including immune cell interactions, physical characteristics, biochemical cues, and metabolomics. Harnessing the advancing knowledge of the FBR allows for the optimization of biomaterial-based implants, aiming to prevent damage of the implant, improve natural regeneration, and provide the tools for an efficient and successful in vivo implantation.
Collapse
Affiliation(s)
- Tim ten Brink
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Febriyani Damanik
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Joris I. Rotmans
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333ZAThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
11
|
T Cruz J, Karen Álvarez, H Orozco V, Mauricio Rojas, A Morales-Luckie R, F Giraldo L. PLGA-LEC/F127 hybrid nanoparticles loaded with curcumin and their modulatory effect on monocytes. Nanomedicine (Lond) 2024; 19:1407-1423. [PMID: 38920352 PMCID: PMC11382718 DOI: 10.1080/17435889.2024.2357530] [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: 03/14/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
Aim: To investigate the effect of surfactant type on curcumin-loaded (CUR) PLGA nanoparticles (NPs) to modulate monocyte functions. Materials & methods: The nanoprecipitation method was used, and PLGA NPs were designed using Pluronic F127 (F127) and/or lecithin (LEC) as surfactants. Results: The Z-average of the NPs was <200 nm, they had a spherical shape, Derjaguin-Muller-Toporov modulus >0.128 MPa, they were stable during storage at 4°C, ζ-potential ∼-40 mV, polydispersity index <0.26 and % EE of CUR >94%. PLGA-LEC/F127 NPs showed favorable physicochemical and nanomechanical properties. These NPs were bound and internalized mainly by monocytes, suppressed monocyte-induced reactive oxygen species production, and decreased the ability of monocytes to modulate T-cell proliferation. Conclusion: These results demonstrate the potential of these NPs for targeted therapy.
Collapse
Affiliation(s)
- Jennifer T Cruz
- Polymer Research Laboratory (LIPOL), Institute of Chemistry, University of Antioquia (UdeA), Medellín, Colombia
- Faculty of Basic Sciences, University of the Amazonia (UDLA), Florencia, Colombia
| | - Karen Álvarez
- Cellular Immunology & Immunogenetics Group (GICIG), University Research Headquarters (SIU), University of Antioquia (UdeA), Medellín, Colombia
| | - Víctor H Orozco
- Polymer Research Laboratory (LIPOL), Institute of Chemistry, University of Antioquia (UdeA), Medellín, Colombia
| | - Mauricio Rojas
- Cellular Immunology & Immunogenetics Group (GICIG), University Research Headquarters (SIU), University of Antioquia (UdeA), Medellín, Colombia
| | - Raul A Morales-Luckie
- Autonomous University of the State of Mexico, Sustainable Chemistry Research Joint Center UAEM-UNAM (CCIQS), Toluca, Estado de México, México
| | - Luis F Giraldo
- Polymer Research Laboratory (LIPOL), Institute of Chemistry, University of Antioquia (UdeA), Medellín, Colombia
| |
Collapse
|
12
|
Zhang Z, Ou L, Yang K, Yuan B. Energy and Speed Landscapes of the Membrane Internalization Behavior of Soft Nanoparticles. J Phys Chem B 2024; 128:2632-2639. [PMID: 38467492 DOI: 10.1021/acs.jpcb.3c07177] [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: 03/13/2024]
Abstract
The cellular endocytosis of nanoparticles (NPs) is a fundamental biological process with significant potential in biomedical applications. However, a comprehensive understanding of the mechanistic aspects of endocytosis and the impact of particle properties on this process remains elusive. In this study, we investigated the membrane-wrapping behavior of soft NPs (SNPs) with varying rigidities using theoretical calculations. Our findings reveal that the membrane-wrapping process of SNPs involves a complex energy change including the possible existence of an energy barrier; moreover, it is found that the location and height of this barrier strongly depend on the mechanistic properties of the NPs and membranes. Additionally, by considering force balance in the membrane-wrapping process, we calculated the speed at which NP is internalized by the membrane, showing a nonmonotonic dependence on particle rigidity and/or wrapping degree. These phenomena can be attributed to competition between different energy components associated with NP-membrane binding, membrane tension, and deformations occurring during SNP-membrane interaction processes. Our results contribute to a deeper understanding of cellular-level endocytosis mechanisms and offer potential applications for soft NPs in biomedicine.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu,China
| | - Luping Ou
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu,China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu,China
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong,China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong,China
| |
Collapse
|
13
|
Mittelheisser V, Gensbittel V, Bonati L, Li W, Tang L, Goetz JG. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. NATURE NANOTECHNOLOGY 2024; 19:281-297. [PMID: 38286876 DOI: 10.1038/s41565-023-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/26/2023] [Indexed: 01/31/2024]
Abstract
Disease progression is usually accompanied by changes in the biochemical composition of cells and tissues and their biophysical properties. For instance, hallmarks of cancer include the stiffening of tissues caused by extracellular matrix remodelling and the softening of individual cancer cells. In this context, accumulating evidence has shown that immune cells sense and respond to mechanical signals from the environment. However, the mechanisms regulating these mechanical aspects of immune surveillance remain partially understood. The growing appreciation for the 'mechano-immunology' field has urged researchers to investigate how immune cells sense and respond to mechanical cues in various disease settings, paving the way for the development of novel engineering strategies that aim at mechanically modulating and potentiating immune cells for enhanced immunotherapies. Recent pioneer developments in this direction have laid the foundations for leveraging 'mechanical immunoengineering' strategies to treat various diseases. This Review first outlines the mechanical changes occurring during pathological progression in several diseases, including cancer, fibrosis and infection. We next highlight the mechanosensitive nature of immune cells and how mechanical forces govern the immune responses in different diseases. Finally, we discuss how targeting the biomechanical features of the disease milieu and immune cells is a promising strategy for manipulating therapeutic outcomes.
Collapse
Affiliation(s)
- Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
14
|
Gaballa SA, Shimizu T, Ando H, Takata H, Emam SE, Ramadan E, Naguib YW, Mady FM, Khaled KA, Ishida T. Treatment-induced and Pre-existing Anti-peg Antibodies: Prevalence, Clinical Implications, and Future Perspectives. J Pharm Sci 2024; 113:555-578. [PMID: 37931786 DOI: 10.1016/j.xphs.2023.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Polyethylene glycol (PEG) is a versatile polymer that is used in numerous pharmaceutical applications like the food industry, a wide range of disinfectants, cosmetics, and many commonly used household products. PEGylation is the term used to describe the covalent attachment of PEG molecules to nanocarriers, proteins and peptides, and it is used to prolong the circulation half-life of the PEGylated products. Consequently, PEGylation improves the efficacy of PEGylated therapeutics. However, after four decades of research and more than two decades of clinical applications, an unappealing side of PEGylation has emerged. PEG immunogenicity and antigenicity are remarkable challenges that confound the widespread clinical application of PEGylated therapeutics - even those under clinical trials - as anti-PEG antibodies (Abs) are commonly reported following the systemic administration of PEGylated therapeutics. Furthermore, pre-existing anti-PEG Abs have also been reported in healthy individuals who have never been treated with PEGylated therapeutics. The circulating anti-PEG Abs, both treatment-induced and pre-existing, selectively bind to PEG molecules of the administered PEGylated therapeutics inducing activation of the complement system, which results in remarkable clinical implications with varying severity. These include increased blood clearance of the administered PEGylated therapeutics through what is known as the accelerated blood clearance (ABC) phenomenon and initiation of serious adverse effects through complement activation-related pseudoallergic reactions (CARPA). Therefore, the US FDA industry guidelines have recommended the screening of anti-PEG Abs, in addition to Abs against PEGylated proteins, in the clinical trials of PEGylated protein therapeutics. In addition, strategies revoking the immunogenic response against PEGylated therapeutics without compromising their therapeutic efficacy are important for the further development of advanced PEGylated therapeutics and drug-delivery systems.
Collapse
Affiliation(s)
- Sherif A Gaballa
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Sherif E Emam
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Eslam Ramadan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Youssef W Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Fatma M Mady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Khaled A Khaled
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
15
|
Agharkar AN, Hajra D, Roy D, Jaiswal V, Kabi P, Chakravortty D, Basu S. Evaporation of bacteria-laden surrogate respiratory fluid droplets: On a hydrophilic substrate vs contact-free environment confers differential bacterial infectivity. PHYSICS OF FLUIDS 2024; 36. [DOI: 10.1063/5.0196219] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The transmission of viruses/bacteria causes infection predominantly via aerosols. The transmission mechanism of respiratory diseases is complex, which includes direct or indirect contact, large droplet, and airborne routes apart from close contact transmission. With this premise, two modes of droplet evaporation are investigated to understand its significance in airborne disease transmission; a droplet in a contact-free environment, which evaporates and forms droplet nuclei, and a droplet on a hydrophilic substrate (fomite). The study examines mass transport, the deposition pattern of bacteria in the precipitates, and their survival and virulence. The osmotic pressure increases with the salt concentration, inactivating the bacteria embedded in the precipitates with accelerated evaporation. Furthermore, the bacteria's degree of survival and enhanced pathogenicity are compared for both evaporation modes. The striking differences in pathogenicity are attributed to the evaporation rate, oxygen availability, and reactive oxygen species generation.
Collapse
Affiliation(s)
- Amey Nitin Agharkar
- Interdisciplinary Centre for Energy Research (ICER), Indian Institute of Science 1 , Bangalore 560012, Karnataka, India
| | - Dipasree Hajra
- Department of Microbiology and Cell Biology, Indian Institute of Science 2 , Bangalore 560012, Karnataka, India
| | - Durbar Roy
- Department of Mechanical Engineering, Indian Institute of Science 3 , Bangalore 560012, Karnataka, India
| | - Vivek Jaiswal
- Department of Mechanical Engineering, Indian Institute of Science 3 , Bangalore 560012, Karnataka, India
| | - Prasenjit Kabi
- Department of Mechanical Engineering, Indian Institute of Science 3 , Bangalore 560012, Karnataka, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science 2 , Bangalore 560012, Karnataka, India
- Adjunct Faculty, School of Biology, Indian Institute of Science Education and Research 4 , Thiruvananthapuram 695551, Kerala, India
| | - Saptarshi Basu
- Interdisciplinary Centre for Energy Research (ICER), Indian Institute of Science 1 , Bangalore 560012, Karnataka, India
- Department of Mechanical Engineering, Indian Institute of Science 3 , Bangalore 560012, Karnataka, India
| |
Collapse
|
16
|
Li Z, Yang X, Li Z. Tumor Mechanics Meets Nanomedicine Mechanical Properties. CHEMISTRY OF MATERIALS 2024; 36:1041-1053. [DOI: 10.1021/acs.chemmater.3c02474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Affiliation(s)
- Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| |
Collapse
|
17
|
Al-Madani H, Yang Y, Refat M, He Q, Peng H, Wu A, Yang F. Quantification and biological evaluation of Zn xFe 3-xO 4 nanoparticle stiffness in a drug delivery system of MCF-7 cancer cells. J Mater Chem B 2024; 12:1636-1651. [PMID: 38270595 DOI: 10.1039/d3tb02723f] [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: 01/26/2024]
Abstract
The delivery of nanoparticles (NPs) to tumors remains challenging despite significant advancements in drug delivery technologies. Addressing this issue requires the establishment of quantitative and reliable criteria to evaluate the cellular absorption of NPs. The mechanical characteristics of NPs and their interaction with cells play a crucial role in cellular drug delivery by influencing cellular internalization. In particular, NPs' stiffness has emerged as a key factor affecting cellular uptake and viability. In this study, we synthesized ZnxFe3-xO4 NPs with varying Zn doping concentrations and conducted an extensive measurement process to investigate the impact of NP stiffness on cellular uptake and the viability of cancerous cells. Initially, the stiffness of the NPs was measured using two methods: single-molecule force spectrometry of atomic force microscopy (SMFS-AFM) and cation distribution as chemical structure analysis. The influence of NP stiffness on intracellular behavior was examined by assessing cellular uptake and viability at different time points during the incubation period. The results obtained from both stiffness measurement methods exhibited consistent trends. NPs with higher stiffness exhibited enhanced cellular uptake but exhibited reduced cellular viability compared to the lower-stiffness NPs. Our findings provide valuable insights into the influence of Zn doping concentration on the mechanical properties of ZnxFe3-xO4 NPs and their consequential impacts on cellular internalization. This study contributes to an improved comprehension of the mechanisms underlying cellular uptake and facilitates advancements in the field of drug transport, thereby enhancing the efficiency of NP-based drug delivery.
Collapse
Affiliation(s)
- Hamzah Al-Madani
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yiqian Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Moath Refat
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Qingxin He
- Guangxi Vocational & Technical Institute of Industry, Guangxi 530001, P. R. China
| | - Hao Peng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China.
| | - Fang Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China.
| |
Collapse
|
18
|
Lu J, Xu X, Sun X, Du Y. Protein and peptide-based renal targeted drug delivery systems. J Control Release 2024; 366:65-84. [PMID: 38145662 DOI: 10.1016/j.jconrel.2023.12.036] [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: 07/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Renal diseases have become an increasingly concerned public health problem in the world. Kidney-targeted drug delivery has profound transformative potential on increasing renal efficacy and reducing extra-renal toxicity. Protein and peptide-based kidney targeted drug delivery systems have garnered more and more attention due to its controllable synthesis, high biocompatibility and low immunogenicity. At the same time, the targeting methods based on protein/peptide are also abundant, including passive renal targeting based on macromolecular protein and active targeting mediated by renal targeting peptide. Here, we review the application and the drug loading strategy of different proteins or peptides in targeted drug delivery, including the ferritin family, albumin, low molecular weight protein (LMWP), different peptide sequence and antibodies. In addition, we summarized the factors influencing passive and active targeting in drug delivery system, the main receptors related to active targeting in different kidney diseases, and a variety of nano forms of proteins based on the controllable synthesis of proteins.
Collapse
Affiliation(s)
- Jingyi Lu
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaoling Xu
- College of Medical Sciences, Zhejiang Shuren University, 8 Shuren Street, Hangzhou, Zhejiang 310015, China.
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China.
| | - Yongzhong Du
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, China.
| |
Collapse
|
19
|
Teer L, Yaddanapudi K, Chen J. Biophysical Control of the Glioblastoma Immunosuppressive Microenvironment: Opportunities for Immunotherapy. Bioengineering (Basel) 2024; 11:93. [PMID: 38247970 PMCID: PMC10813491 DOI: 10.3390/bioengineering11010093] [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: 11/01/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
GBM is the most aggressive and common form of primary brain cancer with a dismal prognosis. Current GBM treatments have not improved patient survival, due to the propensity for tumor cell adaptation and immune evasion, leading to a persistent progression of the disease. In recent years, the tumor microenvironment (TME) has been identified as a critical regulator of these pro-tumorigenic changes, providing a complex array of biomolecular and biophysical signals that facilitate evasion strategies by modulating tumor cells, stromal cells, and immune populations. Efforts to unravel these complex TME interactions are necessary to improve GBM therapy. Immunotherapy is a promising treatment strategy that utilizes a patient's own immune system for tumor eradication and has exhibited exciting results in many cancer types; however, the highly immunosuppressive interactions between the immune cell populations and the GBM TME continue to present challenges. In order to elucidate these interactions, novel bioengineering models are being employed to decipher the mechanisms of immunologically "cold" GBMs. Additionally, these data are being leveraged to develop cell engineering strategies to bolster immunotherapy efficacy. This review presents an in-depth analysis of the biophysical interactions of the GBM TME and immune cell populations as well as the systems used to elucidate the underlying immunosuppressive mechanisms for improving current therapies.
Collapse
Affiliation(s)
- Landon Teer
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| | - Kavitha Yaddanapudi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
- Immuno-Oncology Program, Brown Cancer Center, Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Joseph Chen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| |
Collapse
|
20
|
Yu J, Liu Y, Zhang Y, Ran R, Kong Z, Zhao D, Liu M, Zhao W, Cui Y, Hua Y, Gao L, Zhang Z, Yang Y. Smart nanogels for cancer treatment from the perspective of functional groups. Front Bioeng Biotechnol 2024; 11:1329311. [PMID: 38268937 PMCID: PMC10806105 DOI: 10.3389/fbioe.2023.1329311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction: Cancer remains a significant health challenge, with chemotherapy being a critical treatment modality. However, traditional chemotherapy faces limitations due to non-specificity and toxicity. Nanogels, as advanced drug carriers, offer potential for targeted and controlled drug release, improving therapeutic efficacy and reducing side effects. Methods: This review summarizes the latest developments in nanogel-based chemotherapy drug delivery systems, focusing on the role of functional groups in drug loading and the design of smart hydrogels with controlled release mechanisms. We discuss the preparation methods of various nanogels based on different functional groups and their application in cancer treatment. Results: Nanogels composed of natural and synthetic polymers, such as chitosan, alginate, and polyacrylic acid, have been developed for chemotherapy drug delivery. Functional groups like carboxyl, disulfide, and hydroxyl groups play crucial roles in drug encapsulation and release. Smart hydrogels have been engineered to respond to tumor microenvironmental cues, such as pH, redox potential, temperature, and external stimuli like light and ultrasound, enabling targeted drug release. Discussion: The use of functional groups in nanogel preparation allows for the creation of multifunctional nanogels with high drug loading capacity, controllable release, and good targeting. These nanogels have shown promising results in preclinical studies, with enhanced antitumor effects and reduced systemic toxicity compared to traditional chemotherapy. Conclusion: The development of smart nanogels with functional group-mediated drug delivery and controlled release strategies represents a promising direction in cancer therapy. These systems offer the potential for improved patient outcomes by enhancing drug targeting and minimizing adverse effects. Further research is needed to optimize nanogel design, evaluate their safety and efficacy in clinical trials, and explore their potential for personalized medicine.
Collapse
Affiliation(s)
- Jiachen Yu
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yuting Liu
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
- Shenyang Traditional Chinese Medicine Hospital, China Medical University, Shenyang, China
| | - Yingchun Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Rong Ran
- Department of Anesthesia, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Zixiao Kong
- China Medical University, Shenyang, Liaoning, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Minda Liu
- Department of Oral-maxillofacial Head and Neck, Oral Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Lianbo Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yingxin Yang
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
| |
Collapse
|
21
|
Wells M, Mikesh M, Gordon V. Structure-preserving fixation allows scanning electron microscopy to reveal biofilm microstructure and interactions with immune cells. J Microsc 2024; 293:59-68. [PMID: 38098170 PMCID: PMC10764082 DOI: 10.1111/jmi.13252] [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: 08/31/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Pseudomonas aeruginosa is a pathogen that forms robust biofilms which are commonly associated with chronic infections and cannot be successfully cleared by the immune system. Neutrophils, the most common white blood cells, target infections with pathogen-killing mechanisms that are rendered largely ineffective by the protective physicochemical structure of a biofilm. Visualisation of the complex interactions between immune cells and biofilms will advance understanding of how biofilms evade the immune system and could aid in developing treatment methods that promote immune clearance with minimal harm to the host. Scanning electron microscopy (SEM) distinguishes itself as a powerful, high-resolution tool for obtaining strikingly clear and detailed topographical images. However, taking full advantage of SEM's potential for high-resolution imaging requires that the fixation process simultaneously preserve both intricate biofilm architecture and the morphologies and structural signatures characterising neutrophils responses at an infection site. Standard aldehyde-based fixation techniques result in significant loss of biofilm matrix material and morphologies of responding immune cells, thereby obscuring the details of immune interactions with the biofilm matrix. Here we show an improved fixation technique using the cationic dye alcian blue to preserve and visualise neutrophil interactions with the three-dimensional architecture of P. aeruginosa biofilms. We also demonstrate that this technique better preserves structures of biofilms grown from two other bacterial species, Klebsiella pneumoniae and Burkholderia thailandensis.
Collapse
Affiliation(s)
- Marilyn Wells
- Department of Physics, The University of Texas at Austin, Austin, Texas, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, Texas, USA
| | - Michelle Mikesh
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, Texas, USA
| | - Vernita Gordon
- Department of Physics, The University of Texas at Austin, Austin, Texas, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, Texas, USA
- Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, Texas, USA
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
22
|
Davis MA, Cho E, Teplensky MH. Harnessing biomaterial architecture to drive anticancer innate immunity. J Mater Chem B 2023; 11:10982-11005. [PMID: 37955201 DOI: 10.1039/d3tb01677c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Immunomodulation is a powerful therapeutic approach that harnesses the body's own immune system and reprograms it to treat diseases, such as cancer. Innate immunity is key in mobilizing the rest of the immune system to respond to disease and is thus an attractive target for immunomodulation. Biomaterials have widely been employed as vehicles to deliver immunomodulatory therapeutic cargo to immune cells and raise robust antitumor immunity. However, it is key to consider the design of biomaterial chemical and physical structure, as it has direct impacts on innate immune activation and antigen presentation to stimulate downstream adaptive immunity. Herein, we highlight the widespread importance of structure-driven biomaterial design for the delivery of immunomodulatory cargo to innate immune cells. The incorporation of precise structural elements can be harnessed to improve delivery kinetics, uptake, and the targeting of biomaterials into innate immune cells, and enhance immune activation against cancer through temporal and spatial processing of cargo to overcome the immunosuppressive tumor microenvironment. Structural design of immunomodulatory biomaterials will profoundly improve the efficacy of current cancer immunotherapies by maximizing the impact of the innate immune system and thus has far-reaching translational potential against other diseases.
Collapse
Affiliation(s)
- Meredith A Davis
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Ezra Cho
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Michelle H Teplensky
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Department of Materials Science and Engineering, Boston University, Boston, Massachusetts, 02215, USA
| |
Collapse
|
23
|
Van der Meeren L, Efimova I, Demuynck R, Parakhonskiy B, Krysko DV, Skirtach AG. Mechanobiology of Ferroptotic Cancer Cells as a Novel "Eat-Me" Signal: Regulating Efferocytosis through Layer-by-Layer Coating. Adv Healthc Mater 2023; 12:e2301025. [PMID: 37273241 DOI: 10.1002/adhm.202301025] [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: 04/03/2023] [Revised: 05/31/2023] [Indexed: 06/06/2023]
Abstract
The importance of the clearance of dead cells is shown to have a regulatory role for normal tissue homeostasis and for the modulation of immune responses. However, how mechanobiological properties of dead cells affect efferocytosis remains largely unknown. Here, it is reported that the Young's modulus of cancer cells undergoing ferroptosis is reduced. To modulate their Young's modulus a layer-by-layer (LbL) nanocoating is developed. Scanning electron and fluorescence microscopy confirm coating efficiency of ferroptotic cells while atomic force microscopy reveals encapsulation of the dead cells increases their Young's modulus dependent on the number of applied LbL layers which increases their efferocytosis by primary macrophages. This work demonstrates the crucial role of mechanobiology of dead cells in regulating their efferocytosis by macrophages which can be exploited for the development of novel therapeutic strategies for diseases where modulation of efferocytosis can be potentially beneficial and for the design of drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Louis Van der Meeren
- Nano-BioTechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
| | - Iuliia Efimova
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Robin Demuynck
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Bogdan Parakhonskiy
- Nano-BioTechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
| | - Dmitri V Krysko
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, 9000, Belgium
| | - Andre G Skirtach
- Nano-BioTechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium
| |
Collapse
|
24
|
Montizaan D, Saunders C, Yang K, Sasidharan S, Maity S, Reker-Smit C, Stuart MCA, Montis C, Berti D, Roos WH, Salvati A. Role of Curvature-Sensing Proteins in the Uptake of Nanoparticles with Different Mechanical Properties. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303267. [PMID: 37236202 DOI: 10.1002/smll.202303267] [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: 04/18/2023] [Indexed: 05/28/2023]
Abstract
Nanoparticles of different properties, such as size, charge, and rigidity, are used for drug delivery. Upon interaction with the cell membrane, because of their curvature, nanoparticles can bend the lipid bilayer. Recent results show that cellular proteins capable of sensing membrane curvature are involved in nanoparticle uptake; however, no information is yet available on whether nanoparticle mechanical properties also affect their activity. Here liposomes and liposome-coated silica are used as a model system to compare uptake and cell behavior of two nanoparticles of similar size and charge, but different mechanical properties. High-sensitivity flow cytometry, cryo-TEM, and fluorescence correlation spectroscopy confirm lipid deposition on the silica. Atomic force microscopy is used to quantify the deformation of individual nanoparticles at increasing imaging forces, confirming that the two nanoparticles display distinct mechanical properties. Uptake studies in HeLa and A549 cells indicate that liposome uptake is higher than for the liposome-coated silica. RNA interference studies to silence their expression show that different curvature-sensing proteins are involved in the uptake of both nanoparticles in both cell types. These results confirm that curvature-sensing proteins have a role in nanoparticle uptake, which is not restricted to harder nanoparticles, but includes softer nanomaterials commonly used for nanomedicine applications.
Collapse
Affiliation(s)
- Daphne Montizaan
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Catherine Saunders
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Keni Yang
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Sajitha Sasidharan
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, The Netherlands
| | - Sourav Maity
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, The Netherlands
| | - Catharina Reker-Smit
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Marc C A Stuart
- Electron Microscopy, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, 9747 AG, The Netherlands
| | - Costanza Montis
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, via della Lastruccia 3, Sesto Fiorentino, Florence, 50019, Italy
| | - Debora Berti
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, via della Lastruccia 3, Sesto Fiorentino, Florence, 50019, Italy
| | - Wouter H Roos
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, The Netherlands
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
25
|
Wilson HM. Modulation of macrophages by biophysical cues in health and beyond. DISCOVERY IMMUNOLOGY 2023; 2:kyad013. [PMID: 38567062 PMCID: PMC10917218 DOI: 10.1093/discim/kyad013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/13/2023] [Accepted: 08/09/2023] [Indexed: 04/04/2024]
Abstract
Macrophages play a key role in tissue development and homeostasis, innate immune defence against microbes or tumours, and restoring homeostasis through tissue regeneration following infection or injury. The ability to adopt such diverse functions is due to their heterogeneous nature, which is driven largely by their developmental origin and their response to signals they encounter from the microenvironment. The most well-characterized signals driving macrophage phenotype and function are biochemical and metabolic. However, the way macrophages sense and respond to their extracellular biophysical environment is becoming increasingly recognized in the field of mechano-immunology. These biophysical cues can be signals from tissue components, such as the composition and charge of extracellular matrix or topography, elasticity, and stiffness of the tissue surrounding cells; and mechanical forces such as shear stress or stretch. Macrophages are important in determining whether a disease resolves or becomes chronic. Ageing and diseases such as cancer or fibrotic disorders are associated with significant changes in the tissue biophysical environment, and this provides signals that integrate with those from biochemical and metabolic stimuli to ultimately dictate the overall function of macrophages. This review provides a brief overview of macrophage polarization, followed by a selection of commonly recognized physiological and applied biophysical stimuli impacting macrophage activity, and the potential signalling mechanisms driving downstream responses. The effects of biophysical cues on macrophages' function in homeostasis and disease and the associated clinical implications are also highlighted.
Collapse
Affiliation(s)
- Heather M Wilson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
26
|
Grey EL, McClendon J, Suresh J, Alper S, Janssen WJ, Bryant SJ. Thiol-Michael Addition Microparticles: Their Synthesis, Characterization, and Uptake by Macrophages. ACS Biomater Sci Eng 2023; 9:4223-4240. [PMID: 37379254 PMCID: PMC10619202 DOI: 10.1021/acsbiomaterials.3c00441] [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] [Indexed: 06/30/2023]
Abstract
Polymeric microparticles are promising biomaterial platforms for targeting macrophages in the treatment of disease. This study investigates microparticles formed by a thiol-Michael addition step-growth polymerization reaction with tunable physiochemical properties and their uptake by macrophages. The hexafunctional thiol monomer dipentaerythritol hexa-3-mercaptopropionate (DPHMP) and tetrafunctional acrylate monomer di(trimethylolpropane) tetraacrylate (DTPTA) were reacted in a stepwise dispersion polymerization, achieving tunable monodisperse particles over a size range (1-10 μm) relevant for targeting macrophages. An off-stoichiometry thiol-acrylate reaction afforded facile secondary chemical functionalization to create particles with different chemical moieties. Uptake of the microparticles by RAW 264.7 macrophages was highly dependent on treatment time, particle size, and particle chemistry with amide, carboxyl, and thiol terminal chemistries. The amide-terminated particles were non-inflammatory, while the carboxyl- and thiol-terminated particles induced pro-inflammatory cytokine production in conjunction with particle phagocytosis. Finally, a lung-specific application was explored through time-dependent uptake of amide-terminated particles by human alveolar macrophages in vitro and mouse lungs in vivo without inducing inflammation. The findings demonstrate a promising microparticulate delivery vehicle that is cyto-compatible, is non-inflammatory, and exhibits high rates of uptake by macrophages.
Collapse
Affiliation(s)
- Emerson L. Grey
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Jazalle McClendon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Joshita Suresh
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Scott Alper
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - William J. Janssen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO 80045, USA
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
- Materials Science & Engineering Program, University of Colorado, 4001 Discovery Dr, Boulder, CO 80309-0613, USA
- BioFrontiers Institute, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| |
Collapse
|
27
|
Tischenko K, Brill-Karniely Y, Steinberg E, Segev-Yekutiel H, Benny O. Surface physical cues mediate the uptake of foreign particles by cancer cells. APL Bioeng 2023; 7:016113. [PMID: 36960390 PMCID: PMC10030191 DOI: 10.1063/5.0138245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/22/2023] Open
Abstract
Cancer phenotypes are often associated with changes in the mechanical states of cells and their microenvironments. Numerous studies have established correlations between cancer cell malignancy and cell deformability at the single-cell level. The mechanical deformation of cells is required for the internalization of large colloidal particles. Compared to normal epithelial cells, cancer cells show higher capacities to distort their shapes during the engulfment of external particles, thus performing phagocytic-like processes more efficiently. This link between cell deformability and particle uptake suggests that the cell's adherence state may affect this particle uptake, as cells become stiffer when plated on a more rigid substrate and vice versa. Based on this, we hypothesized that cancer cells of the same origin, which are subjected to external mechanical cues through attachment to surfaces with varying rigidities, may express different capacities to uptake foreign particles. The effects of substrate rigidity on cancer cell uptake of inert particles (0.8 and 2.4 μm) were examined using surfaces with physiologically relevant rigidities (from 0.5 to 64 kPa). Our data demonstrate a wave-like ("meandering") dependence of cell uptake on the rigidity of the culture substrate explained by a superposition of opposing physical and biological effects. The uptake patterns were inversely correlated with the expression of phosphorylated paxillin, indicating that the initial passive particle absorbance is the primary limiting step toward complete uptake. Overall, our findings may provide a foundation for mechanical rationalization of particle uptake design.
Collapse
Affiliation(s)
- Katerina Tischenko
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Yifat Brill-Karniely
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Eliana Steinberg
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Hadas Segev-Yekutiel
- The Core Research Facility, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Ofra Benny
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| |
Collapse
|
28
|
Wells M, Schneider R, Bhattarai B, Currie H, Chavez B, Christopher G, Rumbaugh K, Gordon V. Perspective: The viscoelastic properties of biofilm infections and mechanical interactions with phagocytic immune cells. Front Cell Infect Microbiol 2023; 13:1102199. [PMID: 36875516 PMCID: PMC9978752 DOI: 10.3389/fcimb.2023.1102199] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/24/2023] [Indexed: 02/18/2023] Open
Abstract
Biofilms are viscoelastic materials that are a prominent public health problem and a cause of most chronic bacterial infections, in large part due to their resistance to clearance by the immune system. Viscoelastic materials combine both solid-like and fluid-like mechanics, and the viscoelastic properties of biofilms are an emergent property of the intercellular cohesion characterizing the biofilm state (planktonic bacteria do not have an equivalent property). However, how the mechanical properties of biofilms are related to the recalcitrant disease that they cause, specifically to their resistance to phagocytic clearance by the immune system, remains almost entirely unstudied. We believe this is an important gap that is ripe for a large range of investigations. Here we present an overview of what is known about biofilm infections and their interactions with the immune system, biofilm mechanics and their potential relationship with phagocytosis, and we give an illustrative example of one important biofilm-pathogen (Pseudomonas aeruginosa) which is the most-studied in this context. We hope to inspire investment and growth in this relatively-untapped field of research, which has the potential to reveal mechanical properties of biofilms as targets for therapeutics meant to enhance the efficacy of the immune system.
Collapse
Affiliation(s)
- Marilyn Wells
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, United States
| | - Rebecca Schneider
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Bikash Bhattarai
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, United States
| | - Hailey Currie
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, United States
| | - Bella Chavez
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, United States
| | - Gordon Christopher
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, United States
| | - Kendra Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Vernita Gordon
- Department of Physics, Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, United States
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX, United States
- Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
29
|
Yu Y, Li S, Yao Y, Shen X, Li L, Huang Y. Increasing stiffness promotes pulmonary retention of ligand-directed dexamethasone-loaded nanoparticle for enhanced acute lung inflammation therapy. Bioact Mater 2023; 20:539-547. [PMID: 35846844 PMCID: PMC9253482 DOI: 10.1016/j.bioactmat.2022.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Inhaled nanoparticles (NPs) need to penetrate the bronchial mucosa to deliver drug payloads deeply in the lung for amplified local therapy. However, the bronchial mucociliary barrier eliminates NPs rapidly, which considerably limits their mucosal penetration. In this study, we find that surface ligand modification and stiffness adjustment of NPs contribute to the significantly enhanced bronchial mucosal absorption and pulmonary retention of inhaled drugs. We utilize neonatal Fc receptor ligand (FcBP) to modify the rationally designed low stiffness NPs (Soft-NP) and high stiffness NPs (Stiff-NP) to target bronchial mucosa. In an acute lung inflammation rat model, after intranasal administration with dexamethasone-loaded NPs, Stiff-NP endowed with FcBP displays superior therapeutic effects. The in vitro data demonstrate that the promotion effect of FcBP to bronchial mucosal absorption of Stiff-NP dominates over Soft-NP. This could be attributed to the higher affinity between ligand-receptor when incorporating FcBP on the Stiff-NP surface. Meanwhile, high stiffness modulates more actin filaments aggregation to mediate endocytosis, along with strengthened Ca2+ signal to enhance exocytosis. Conclusively, we highlight that FcBP-modified NPs with higher stiffness would be a potential pulmonary drug delivery system.
Collapse
Affiliation(s)
- Yinglan Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shujie Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuan Yao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xinran Shen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
30
|
Dols-Perez A, Fornaguera C, Feiner-Gracia N, Grijalvo S, Solans C, Gomila G. Effect of surface functionalization and loading on the mechanical properties of soft polymeric nanoparticles prepared by nano-emulsion templating. Colloids Surf B Biointerfaces 2023; 222:113019. [PMID: 36435028 DOI: 10.1016/j.colsurfb.2022.113019] [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: 07/29/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022]
Abstract
Drug and gene delivery systems based on polymeric nanoparticles offer a greater efficacy and a reduced toxicity compared to traditional formulations. Recent studies have evidenced that their internalization, biodistribution and efficacy can be affected, among other factors, by their mechanical properties. Here, we analyze by means of Atomic Force Microscopy force spectroscopy how composition, surface functionalization and loading affect the mechanics of nanoparticles. For this purpose, nanoparticles made of Poly(lactic-co-glycolic) (PLGA) and Ethyl cellulose (EC) with different functionalizations and loading were prepared by nano-emulsion templating using the Phase Inversion Composition method (PIC) to form the nano-emulsions. A multiparametric nanomechanical study involving the determination of the Young's modulus, maximum deformation and breakthrough force was carried out. The obtained results showed that composition, surface functionalization and loading affect the nanomechanical properties in a different way, thus requiring, in general, to consider the overall mechanical properties after the addition of a functionalization or loading. A graphical representation method has been proposed enabling to easily identify mechanically equivalent formulations, which is expected to be useful in the development of soft polymeric nanoparticles for pre-clinical and clinical use.
Collapse
Affiliation(s)
- Aurora Dols-Perez
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), C/Jordi Girona 18-26, 08034 Barcelona, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; Institut de Bioenginyeria de Catalunya (IBEC), C/ Balidiri i Reixac 15-21, 08028 Barcelona, Spain; Departament of Electronics and Biomedical Engineering, Universitat de Barcelona, C/ Martí i Franquès 1, 08028 Barcelona, Spain.
| | - Cristina Fornaguera
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), C/Jordi Girona 18-26, 08034 Barcelona, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; Grup d'Enginyeria de Materials (Gemat) - Institut Químic de Sarrià (IQS) - Universitat Ramon Llull (URL), Barcelona, Spain
| | - Natalia Feiner-Gracia
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), C/Jordi Girona 18-26, 08034 Barcelona, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Santiago Grijalvo
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), C/Jordi Girona 18-26, 08034 Barcelona, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Conxita Solans
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), C/Jordi Girona 18-26, 08034 Barcelona, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Gabriel Gomila
- Institut de Bioenginyeria de Catalunya (IBEC), C/ Balidiri i Reixac 15-21, 08028 Barcelona, Spain; Departament of Electronics and Biomedical Engineering, Universitat de Barcelona, C/ Martí i Franquès 1, 08028 Barcelona, Spain
| |
Collapse
|
31
|
The Effect of Elasticity of Gelatin Nanoparticles on the Interaction with Macrophages. Pharmaceutics 2023; 15:pharmaceutics15010199. [PMID: 36678828 PMCID: PMC9861130 DOI: 10.3390/pharmaceutics15010199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Gelatin is a biocompatible, biodegradable, cheap, and nontoxic material, which is already used for pharmaceutical applications. Nanoparticles from gelatin (GNPs) are considered a promising delivery system for hydrophilic and macromolecular drugs. Mechanical properties of particles are recognized as an important parameter affecting drug carrier interaction with biological systems. GNPs offer the preparation of particles with different stiffness. GNPs were loaded with Fluorescein isothiocyanate-labeled 150 kDa dextran (FITC-dextran) yielding also different elastic properties. GNPs were visualized using atomic force microscopy (AFM), and force-distance curves from the center of the particles were evaluated for Young's modulus calculation. The prepared GNPs have Young's moduli from 4.12 MPa for soft to 9.8 MPa for stiff particles. Furthermore, cytokine release (IL-6 and TNF-α), cell viability, and cell uptake were determined on macrophage cell lines from mouse (RAW 264.7) and human (dTHP-1 cells, differentiated human monocytic THP-1 cells) origin for soft and stiff GNPs. Both particle types showed good cell compatibility and did not induce IL-6 and TNF-α release from RAW 264.7 and dTHP-1 cells. Stiffer GNPs were internalized into cells faster and to a larger extent.
Collapse
|
32
|
Shi J, Wu X, Wang Z, Li F, Meng Y, Moore RM, Cui J, Xue C, Croce KR, Yurdagul A, Doench JG, Li W, Zarbalis KS, Tabas I, Yamamoto A, Zhang H. A genome-wide CRISPR screen identifies WDFY3 as a regulator of macrophage efferocytosis. Nat Commun 2022; 13:7929. [PMID: 36566259 PMCID: PMC9789999 DOI: 10.1038/s41467-022-35604-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
Phagocytic clearance of dying cells, termed efferocytosis, is essential for maintaining tissue homeostasis, yet our understanding of efferocytosis regulation remains incomplete. Here we perform a FACS-based, genome-wide CRISPR knockout screen in primary mouse macrophages to search for novel regulators of efferocytosis. The results show that Wdfy3 knockout in macrophages specifically impairs uptake, but not binding, of apoptotic cells due to defective actin disassembly. Additionally, WDFY3 interacts with GABARAP, thus facilitating LC3 lipidation and subsequent lysosomal acidification to permit the degradation of apoptotic cell components. Mechanistically, while the C-terminus of WDFY3 is sufficient to rescue the impaired degradation induced by Wdfy3 knockout, full-length WDFY3 is required to reconstitute the uptake of apoptotic cells. Finally, WDFY3 is also required for efficient efferocytosis in vivo in mice and in vitro in primary human macrophages. This work thus expands our knowledge of the mechanisms of macrophage efferocytosis, as well as supports genome-wide CRISPR screen as a platform for interrogating complex functional phenotypes in primary macrophages.
Collapse
Affiliation(s)
- Jianting Shi
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ziyi Wang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Fang Li
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Yujiao Meng
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Beijing University of Chinese Medicine, Beijing, China
| | - Rebecca M Moore
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jian Cui
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Chenyi Xue
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Katherine R Croce
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Arif Yurdagul
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wei Li
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
| | - Konstantinos S Zarbalis
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA
- UC Davis MIND Institute, Sacramento, CA, 95817, USA
| | - Ira Tabas
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Ai Yamamoto
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
33
|
Barger SR, Vorselen D, Gauthier NC, Theriot JA, Krendel M. F-actin organization and target constriction during primary macrophage phagocytosis is balanced by competing activity of myosin-I and myosin-II. Mol Biol Cell 2022; 33:br24. [PMID: 36129777 PMCID: PMC9727803 DOI: 10.1091/mbc.e22-06-0210] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Phagocytosis requires rapid remodeling of the actin cytoskeleton for extension of membrane protrusions and force generation to ultimately drive the engulfment of targets. The detailed mechanisms of phagocytosis have almost exclusively been studied in immortalized cell lines. Here, we make use of high-resolution imaging and novel biophysical approaches to determine the structural and mechanical features of phagocytosis by primary bone marrow-derived macrophages. We find that the signature behavior of these primary cells is distinct from macrophage-like cell lines; specifically, it is gentle, with only weak target constriction and modest polarization of the F-actin distribution inside the phagocytic cup. We show that long-tailed myosins 1e/f are critical for this organization. Deficiency of myo1e/f causes dramatic shifts in F-actin localization, reducing F-actin at the phagocytic cup base and enhancing F-actin-mediated constriction at the cup rim. Surprisingly, these changes can be almost fully reverted upon inhibition of another myosin motor protein, myosin-II. Hence, we show that the biomechanics and large-scale organization of phagocytic cups is tightly regulated through competing contributions from myosin-Ie/f and myosin-II.
Collapse
Affiliation(s)
- Sarah R Barger
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Daan Vorselen
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195.,Cell Biology and Immunology, Wageningen University and Research, Wageningen, the Netherlands
| | | | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
34
|
Vorselen D. Dynamics of phagocytosis mediated by phosphatidylserine. Biochem Soc Trans 2022; 50:1281-1291. [PMID: 36281986 PMCID: PMC9704538 DOI: 10.1042/bst20211254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 10/13/2023]
Abstract
Phagocytosis triggered by the phospholipid phosphatidylserine (PS) is key for the removal of apoptotic cells in development, tissue homeostasis and infection. Modulation of PS-mediated phagocytosis is an attractive target for therapeutic intervention in the context of atherosclerosis, neurodegenerative disease, and cancer. Whereas the mechanisms of target recognition, lipid and protein signalling, and cytoskeletal remodelling in opsonin-driven modes of phagocytosis are increasingly well understood, PS-mediated phagocytosis has remained more elusive. This is partially due to the involvement of a multitude of receptors with at least some redundancy in functioning, which complicates dissecting their contributions and results in complex downstream signalling networks. This review focusses on the receptors involved in PS-recognition, the signalling cascades that connect receptors to cytoskeletal remodelling required for phagocytosis, and recent progress in our understanding of how phagocytic cup formation is coordinated during PS-mediated phagocytosis.
Collapse
Affiliation(s)
- Daan Vorselen
- Department of Biology, University of Washington, Seattle, WA 98105, U.S.A
| |
Collapse
|
35
|
Lin H, Zhang Y, Dong S, Cai X, Jiang H, Fan Y, Ying K, Du B, Yu P, Yang W. Targeted Therapy of Ischemic Stroke via Crossing the Blood-Brain Barrier Using Edaravone-Loaded Multiresponsive Microgels. ACS APPLIED BIO MATERIALS 2022; 5:4165-4178. [PMID: 36083038 DOI: 10.1021/acsabm.2c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemic stroke, as a prevalent neurological disorder, often results in rapid increases in the production of reactive oxygen species (ROS) and inflammatory factors in the focal ischemic area. Though edaravone is an approved treatment, its effect is limited due to its weak ability to cross the blood-brain barrier (BBB) and short half-life. Other effective pharmacological treatment options are clearly lacking. In this study, PNIVDBrF-3-Eda (also named MG-3-Eda) was prepared using a thermo- and pH dual-responsive PNIVDBrF microgel. These were designed with a positively charged network, as synthesized by simultaneous quaternization cross-linking and surfactant-free emulsion copolymerization, to be loaded with the negatively charged edaravone. We then investigated whether such a targeted delivery of edaravone could provide enhanced neuroprotection. Cytotoxicity assays confirmed that the microgel (<1 mg/mL) exhibited promising cytocompatibility with no remarkable effect on cell viability, cell cycle regulation, or apoptosis levels. In vitro and in vivo experiments demonstrated that the microgels could successfully penetrate the blood-brain barrier where efficient BBB crossing was observed after disruption of the BBB due to ischemic injury. This enabled MG-3-Eda to target the cerebral ischemic area and achieve local release of edaravone. Treatment with MG-3-Eda significantly reduced the cerebral infarct area in transient middle cerebral artery occlusion (tMCAO) mice and significantly improved behavioral scores. MG-3-Eda treatment also prevented the reduction in NF200 expression, a neuronal marker protein, and attenuated microglia activation (as detected by Iba1) in the local ischemic area via local antioxidant and anti-inflammatory effects. A superior neuroprotective effect was noted for MG-3-Eda compared to that for free edaravone. Our results indicate that MG-3-Eda administration represents a clear potential treatment for cerebral ischemia via its targeted delivery of edaravone to ischemic areas where it provides significant local antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Hongwei Lin
- Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yi Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Xiaobo Cai
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hui Jiang
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yang Fan
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Kaiyue Ying
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Peilin Yu
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
36
|
Kalashnikov N, Moraes C. Engineering physical microenvironments to study innate immune cell biophysics. APL Bioeng 2022; 6:031504. [PMID: 36156981 PMCID: PMC9492295 DOI: 10.1063/5.0098578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Innate immunity forms the core of the human body's defense system against infection, injury, and foreign objects. It aims to maintain homeostasis by promoting inflammation and then initiating tissue repair, but it can also lead to disease when dysregulated. Although innate immune cells respond to their physical microenvironment and carry out intrinsically mechanical actions such as migration and phagocytosis, we still do not have a complete biophysical description of innate immunity. Here, we review how engineering tools can be used to study innate immune cell biophysics. We first provide an overview of innate immunity from a biophysical perspective, review the biophysical factors that affect the innate immune system, and then explore innate immune cell biophysics in the context of migration, phagocytosis, and phenotype polarization. Throughout the review, we highlight how physical microenvironments can be designed to probe the innate immune system, discuss how biophysical insight gained from these studies can be used to generate a more comprehensive description of innate immunity, and briefly comment on how this insight could be used to develop mechanical immune biomarkers and immunomodulatory therapies.
Collapse
Affiliation(s)
- Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | | |
Collapse
|
37
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
38
|
Li M, Jin X, Liu T, Fan F, Gao F, Chai S, Yang L. Nanoparticle elasticity affects systemic circulation lifetime by modulating adsorption of apolipoprotein A-I in corona formation. Nat Commun 2022; 13:4137. [PMID: 35842431 PMCID: PMC9288426 DOI: 10.1038/s41467-022-31882-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Nanoparticle elasticity is crucial in nanoparticles' physiological fate, but how this occurs is largely unknown. Using core-shell nanoparticles with a same PEGylated lipid bilayer shell yet cores differing in elasticity (45 kPa - 760 MPa) as models, we isolate the effects of nanoparticle elasticity from those of other physiochemical parameters and, using mouse models, observe a non-monotonic relationship of systemic circulation lifetime versus nanoparticle elasticity. Incubating our nanoparticles in mouse plasma provides protein coronas varying non-monotonically in composition depending on nanoparticle elasticity. Particularly, apolipoprotein A-I (ApoA1) is the only protein whose relative abundance in corona strongly correlates with our nanoparticles' blood clearance lifetime. Notably, similar results are observed when above nanoparticles' PEGylated lipid bilayer shell is changed to be non-PEGylated. This work unveils the mechanisms by which nanoparticle elasticity affects nanoparticles' physiological fate and suggests nanoparticle elasticity as a readily tunable parameter in future rational exploiting of protein corona.
Collapse
Affiliation(s)
- Mingyang Li
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xinyang Jin
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Tao Liu
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Feng Fan
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Feng Gao
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Shuang Chai
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China.,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Lihua Yang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, Anhui, China. .,CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, Anhui, China. .,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, Anhui, China.
| |
Collapse
|
39
|
Garcia‐Herreros A, Yeh Y, Peng Z, del Álamo JC. Cyclic Mechanical Stresses Alter Erythrocyte Membrane Composition and Microstructure and Trigger Macrophage Phagocytosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201481. [PMID: 35508805 PMCID: PMC9284186 DOI: 10.1002/advs.202201481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Indexed: 06/01/2023]
Abstract
Red blood cells (RBCs) are cleared from the circulation when they become damaged or display aging signals targeted by macrophages. This process occurs mainly in the spleen, where blood flows through submicrometric constrictions called inter-endothelial slits (IES), subjecting RBCs to large-amplitude deformations. In this work, RBCs are circulated through microfluidic devices containing microchannels that replicate the IES. The cyclic mechanical stresses experienced by the cells affect their biophysical properties and molecular composition, accelerating cell aging. Specifically, RBCs quickly transition to a more spherical, less deformable phenotype that hinders microchannel passage, causing hemolysis. This transition is associated with the release of membrane vesicles, which self-extinguishes as the spacing between membrane-cytoskeleton linkers becomes tighter. Proteomics analysis of the mechanically aged RBCs reveals significant losses of essential proteins involved in antioxidant protection, gas transport, and cell metabolism. Finally, it is shown that these changes make mechanically aged RBCs more susceptible to macrophage phagocytosis. These results provide a comprehensive model explaining how physical stress induces RBC clearance in the spleen. The data also suggest new biomarkers of early "hemodamage" and inflammation preceding hemolysis in RBCs subjected to mechanical stress.
Collapse
Affiliation(s)
- Antoni Garcia‐Herreros
- Department of Mechanical and Aerospace EngineeringUniversity of California9500 Gilman Dr.La JollaCA92093USA
| | - Yi‐Ting Yeh
- Department of Mechanical and Aerospace EngineeringUniversity of California9500 Gilman Dr.La JollaCA92093USA
- Department of BioengineeringUniversity of California9500 Gilman Dr.La JollaCA92093USA
- Institute of Engineering in MedicineUniversity of California9500 Gilman Dr.La JollaCA92093USA
| | - Zhangli Peng
- Department of BioengineeringUniversity of Illinois at Chicago1200 W Harrison StChicagoIL60607USA
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace EngineeringUniversity of California9500 Gilman Dr.La JollaCA92093USA
- Institute of Engineering in MedicineUniversity of California9500 Gilman Dr.La JollaCA92093USA
- Department of Mechanical EngineeringUniversity of Washington850 Republican StSeattleWA98109USA
- Center for Cardiovascular BiologyUniversity of Washington850 Republican StSeattleWA98109USA
| |
Collapse
|
40
|
Buffer Components Incorporate into the Framework of Polyserotonin Nanoparticles and Films during Synthesis. NANOMATERIALS 2022; 12:nano12122027. [PMID: 35745365 PMCID: PMC9227592 DOI: 10.3390/nano12122027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
Polyserotonin nanoparticles (PSeNP) and films are bioinspired nanomaterials that have potential in biomedical applications and surface coatings. As studies on polyserotonin (PSe) nanoparticles and films are still in their infancy, synthetic pathways and material development for this new class of nanomaterial await investigation. Here, we sought to determine how different buffers used during the polymerization of serotonin to form nanoparticles and films impact the physicochemical properties of PSe materials. We show that buffer components are incorporated into the polymer matrix, which is also supported by density functional theory calculations. While we observed no significant differences between the elasticity of nanoparticles synthesized in the different buffers, the nanoscale surface properties of PSe films revealed dissimilarities in surface functional groups influenced by solvent molecules. Overall, the results obtained in this work can be used towards the rational design of PSe nanomaterials with tailored properties and for specific applications.
Collapse
|
41
|
Iaquinta S, Khazaie S, Ishow É, Blanquart C, Fréour S, Jacquemin F. Influence of the mechanical and geometrical parameters on the cellular uptake of nanoparticles: A stochastic approach. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3598. [PMID: 35343089 DOI: 10.1002/cnm.3598] [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: 11/12/2021] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
Nanoparticles (NPs) are used for drug delivery with enhanced selectivity and reduced side-effect toxicity in cancer treatments. Based on the literature, the influence of the NPs mechanical and geometrical properties on their cellular uptake has been studied through experimental investigations. However, due to the difficulty to vary the parameters independently in such a complex system, it remains hard to efficiently conclude on the influence of each one of them on the cellular internalization of a NP. In this context, different mechanical / mathematical models for the cellular uptake of NPs have been developed. In this paper, we numerically investigate the influence of the NP's aspect ratio, the membrane tension and the cell-NP adhesion on the uptake of the NP using the model introduced in1 coupled with a numerical stochastic scheme to measure the weight of each one of the aforementioned parameters. The results reveal that the aspect ratio of the particle is the most influential parameter on the wrapping of the particle by the cell membrane. Then the adhesion contributes twice as much as the membrane tension. Our numerical results match the previous experimental observations.
Collapse
Affiliation(s)
- Sarah Iaquinta
- Nantes Université, Ecole Centrale Nantes, CNRS, GeM, UMR 6183, Saint-Nazaire, France
| | - Shahram Khazaie
- Nantes Université, Ecole Centrale Nantes, CNRS, GeM, UMR 6183, Saint-Nazaire, France
| | - Éléna Ishow
- Nantes Université, CNRS, CEISAM, UMR 6230, Nantes, France
| | - Christophe Blanquart
- Nantes Université, Univ Angers, CHU Nantes, INSERM, CNRS, CRCI2NA, Nantes, France
| | - Sylvain Fréour
- Nantes Université, Ecole Centrale Nantes, CNRS, GeM, UMR 6183, Saint-Nazaire, France
| | - Frédéric Jacquemin
- Nantes Université, Ecole Centrale Nantes, CNRS, GeM, UMR 6183, Saint-Nazaire, France
| |
Collapse
|
42
|
Benne N, Ter Braake D, Stoppelenburg AJ, Broere F. Nanoparticles for Inducing Antigen-Specific T Cell Tolerance in Autoimmune Diseases. Front Immunol 2022; 13:864403. [PMID: 35392079 PMCID: PMC8981588 DOI: 10.3389/fimmu.2022.864403] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Autoimmune diseases affect many people worldwide. Current treatment modalities focus on the reduction of disease symptoms using anti-inflammatory drugs which can lead to side effects due to systemic immune suppression. Restoration of immune tolerance by down-regulating auto-reactive cells in an antigen-specific manner is currently the “holy grail” for the treatment of autoimmune diseases. A promising strategy is the use of nanoparticles that can deliver antigens to antigen-presenting cells which in turn can enhance antigen-specific regulatory T cells. In this review, we highlight some promising cell targets (e.g. liver sinusoidal endothelial cells and splenic marginal zone macrophages) for exploiting natural immune tolerance processes, and several strategies by which antigen-carrying nanoparticles can target these cells. We also discuss how nanoparticles carrying immunomodulators may be able to activate tolerance in other antigen-presenting cell types. Finally, we discuss some important aspects that must be taken into account when translating data from animal studies to patients.
Collapse
Affiliation(s)
- Naomi Benne
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Daniëlle Ter Braake
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Arie Jan Stoppelenburg
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
43
|
Zhou Y, Wu Y. Substrate Viscoelasticity Amplifies Distinctions between Transient and Persistent LPS-Induced Signals. Adv Healthc Mater 2022; 11:e2102271. [PMID: 34855279 DOI: 10.1002/adhm.202102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/22/2021] [Indexed: 01/18/2023]
Abstract
Macrophages settle in heterogeneous microenvironments rendered by other cells and extracellular matrices. It is well known that chemical stimuli direct macrophage behavior; however, the contributions of viscosity, which increases in inflammatory tissues but not in tumors, are ignored in immune responses including effective activation and timely attenuation. This paper demonstrates that transient lipopolysaccharide (LPS)-treated macrophages benefit from elastic substrates, whereas viscoelastic substrates with similar storage moduli support the inflammatory responses of macrophages under persistent stimulations and consequently amplify the distinctions between the transient and persistent LPS-induced transcriptional programs. Actin filaments (F-actin) fluctuate in line with transcriptional profiles and can be mathematically predicted by a clutch-like model. Moreover, viscosity modifies immune responses through transcription factors NF-κB and C/EBPδ, which act as switches discriminating transient and persistent infections. Interestingly, enhanced immune responses, consistent with the lower activated states, are attenuated promptly by the actin nucleation-related translocation of ATF3 to nuclei. These findings suggest that the substrate viscoelasticity induces more intense inflammation only in the case of persistent infection and promotes more sensitively perceiving the duration of infection through the F-actin correlated transcription factors. In addition, it may facilitate the cognition of immune response in inflammatory and cancerous microenvironments and have a wide range of applications in inflammatory regulations.
Collapse
Affiliation(s)
- Yu‐Wei Zhou
- Department of Engineering Mechanics School of Aeronautics and Astronautics Zhejiang University Hangzhou Zhejiang 310027 China
| | - Yu Wu
- Department of Engineering Mechanics School of Aeronautics and Astronautics Zhejiang University Hangzhou Zhejiang 310027 China
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province Zhejiang University Hangzhou Zhejiang 310027 China
- Soft Matter Research Center Zhejiang University Hangzhou Zhejiang 310027 China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou Zhejiang 310027 China
| |
Collapse
|
44
|
Ding X, Li M, Cheng B, Wei Z, Dong Y, Xu F. Microsphere sensors for characterizing stress fields within three-dimensional extracellular matrix. Acta Biomater 2022; 141:1-13. [PMID: 34979325 DOI: 10.1016/j.actbio.2021.12.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Stress in the three-dimensional extracellular matrix is one of the key cues in regulating multiscale biological processes. Thus far, noticeable progress in methods and techniques (e.g., micropipette aspiration, AFM, and molecule probes) has been made to quantify stress in cell microenvironment at different length scales. Among them, the microsphere sensor-based method (MSS-based method) has emerged as an advantageous approach over conventional techniques in quantifying stress in situ and in vivo at cellular and supra-cellular scales. This method is implemented by seven sequential steps, including fabrication, modification, characterization, cell adhesion, imaging, displacement field extraction and stress calculation. Precise control of each step and inter-tunning between steps can provide quantitative characterization of stress field. However, detailed procedural information associated with each step and process has been scattered. This review aims to provide a comprehensive overview of MSS-based method, systematically summarizing the principles and research progresses. Firstly, the basic principles are introduced, and the specific experiment and calculation processes of MSS-based method are presented in detail. Then, recent advances and applications of this method are summarized. Finally, perspectives of the limitations and development trends of MSS-based method are discussed. This specific and comprehensive review would provide a guideline for the widespread application of MSS-based method as an advantageous method for in situ and in vivo stress characterization at cellular and supra-cellular scale within three-dimensional extracellular matrix. STATEMENT OF SIGNIFICANCE: In this review, a method based on a microsphere sensor (MSS-based method) as an advantageous approach over conventional techniques in quantifying stress in situ and in vivo at cellular and supra-cellular scales is introduced and discussed. This technique is implemented by seven sequential steps, including fabrication, modification, characterization, cell junction, imaging, displacement field extraction, and stress calculation. Precise control of each step and inter-tunning between steps can provide quantitative stress field. However, detailed procedural information associated with each step has been scattered. Thus, a comprehensive review collating recent advances and perspective discussions is a necessity to introduce a better option for quantifying the stress field in biological processes at the cellular and supra-cellular scales.
Collapse
Affiliation(s)
- Xin Ding
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Moxiao Li
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
45
|
Norling K, Sjöberg M, Bally M, Zhdanov VP, Parveen N, Höök F. Dissimilar Deformation of Fluid- and Gel-Phase Liposomes upon Multivalent Interaction with Cell Membrane Mimics Revealed Using Dual-Wavelength Surface Plasmon Resonance. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:2550-2560. [PMID: 35156833 PMCID: PMC8892953 DOI: 10.1021/acs.langmuir.1c03096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The mechanical properties of biological nanoparticles play a crucial role in their interaction with the cellular membrane, in particular for cellular uptake. This has significant implications for the design of pharmaceutical carrier particles. In this context, liposomes have become increasingly popular, among other reasons due to their customizability and easily varied physicochemical properties. With currently available methods, it is, however, not trivial to characterize the mechanical properties of nanoscopic liposomes especially with respect to the level of deformation induced upon their ligand-receptor-mediated interaction with laterally fluid cellular membranes. Here, we utilize the sensitivity of dual-wavelength surface plasmon resonance to probe the size and shape of bound liposomes (∼100 nm in diameter) as a means to quantify receptor-induced deformation during their interaction with a supported cell membrane mimic. By comparing biotinylated liposomes in gel and fluid phases, we demonstrate that fluid-phase liposomes are more prone to deformation than their gel-phase counterparts upon binding to the cell membrane mimic and that, as expected, the degree of deformation depends on the number of ligand-receptor pairs that are engaged in the multivalent binding.
Collapse
Affiliation(s)
- Karin Norling
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Mattias Sjöberg
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Marta Bally
- Department
of Clinical Microbiology, Umeå University, 901 85 Umeå, Sweden
- Wallenberg
Centre for Molecular Medicine, Umeå
University, 901 85 Umeå, Sweden
| | - Vladimir P. Zhdanov
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Boreskov
Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nagma Parveen
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- (N.P.)
| | - Fredrik Höök
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- (F.H.)
| |
Collapse
|
46
|
Zak A, Dupré-Crochet S, Hudik E, Babataheri A, Barakat AI, Nüsse O, Husson J. Distinct timing of neutrophil spreading and stiffening during phagocytosis. Biophys J 2022; 121:1381-1394. [PMID: 35318004 PMCID: PMC9072703 DOI: 10.1016/j.bpj.2022.03.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/29/2021] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Phagocytic cells form the first line of defense in an organism, engulfing microbial pathogens. Phagocytosis involves cell mechanical changes that are not yet well understood. Understanding these mechanical modifications promises to shed light on the immune processes that trigger pathological complications. Previous studies showed that phagocytes undergo a sequence of spreading events around their target followed by an increase in cell tension. Seemingly in contradiction, other studies observed an increase in cell tension concomitant with membrane expansion. Even though phagocytes are viscoelastic, few studies have quantified viscous changes during phagocytosis. It is also unclear whether cell lines behave mechanically similarly to primary neutrophils. We addressed the question of simultaneous versus sequential spreading and mechanical changes during phagocytosis by using immunoglobulin-G-coated 8- and 20-μm-diameter beads as targets. We used a micropipette-based single-cell rheometer to monitor viscoelastic properties during phagocytosis by both neutrophil-like PLB cells and primary human neutrophils. We show that the faster expansion of PLB cells on larger beads is a geometrical effect reflecting a constant advancing speed of the phagocytic cup. Cells become stiffer on 20- than on 8-μm beads, and the relative timing of spreading and stiffening of PLB cells depends on target size: on larger beads, stiffening starts before maximal spreading area is reached but ends after reaching maximal area. On smaller beads, the stiffness begins to increase after cells have engulfed the bead. Similar to PLB cells, primary cells become stiffer on larger beads but start spreading and stiffen faster, and the stiffening begins before the end of spreading on both bead sizes. Our results show that mechanical changes in phagocytes are not a direct consequence of cell spreading and that models of phagocytosis should be amended to account for causes of cell stiffening other than membrane expansion.
Collapse
Affiliation(s)
- Alexandra Zak
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France; Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Sophie Dupré-Crochet
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Elodie Hudik
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Avin Babataheri
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Oliver Nüsse
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Julien Husson
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France.
| |
Collapse
|
47
|
Tello-Lafoz M, de Jesus MM, Huse M. Harder, better, faster, stronger: biochemistry and biophysics in the immunosurveillance concert. Trends Immunol 2022; 43:96-105. [PMID: 34973924 PMCID: PMC8810625 DOI: 10.1016/j.it.2021.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023]
Abstract
Antitumor immunosurveillance is triggered by immune cell recognition of characteristic biochemical signals on the surfaces of cancer cells. Recent data suggest that the mechanical properties of cancer cells influence the strength of these signals, with physically harder target cells (more rigid) eliciting better, faster, and stronger cytotoxic responses against metastasis. Using analogies to a certain electronic music duo, we argue that the biophysical properties of cancer cells and their environment can adjust the volume and tone of the antitumor immune response. We also consider the potential influence of biomechanics-based immunosurveillance in disease progression and posit that targeting the biophysical properties of cancer cells in concert with their biochemical features could increase the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Maria Tello-Lafoz
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel M de Jesus
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
48
|
Russo V, El Khatib M, Prencipe G, Cerveró-Varona A, Citeroni MR, Mauro A, Berardinelli P, Faydaver M, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Liverani L, Boccaccini AR, Barboni B. Scaffold-Mediated Immunoengineering as Innovative Strategy for Tendon Regeneration. Cells 2022; 11:cells11020266. [PMID: 35053383 PMCID: PMC8773518 DOI: 10.3390/cells11020266] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Tendon injuries are at the frontier of innovative approaches to public health concerns and sectoral policy objectives. Indeed, these injuries remain difficult to manage due to tendon’s poor healing ability ascribable to a hypo-cellularity and low vascularity, leading to the formation of a fibrotic tissue affecting its functionality. Tissue engineering represents a promising solution for the regeneration of damaged tendons with the aim to stimulate tissue regeneration or to produce functional implantable biomaterials. However, any technological advancement must take into consideration the role of the immune system in tissue regeneration and the potential of biomaterial scaffolds to control the immune signaling, creating a pro-regenerative environment. In this context, immunoengineering has emerged as a new discipline, developing innovative strategies for tendon injuries. It aims at designing scaffolds, in combination with engineered bioactive molecules and/or stem cells, able to modulate the interaction between the transplanted biomaterial-scaffold and the host tissue allowing a pro-regenerative immune response, therefore hindering fibrosis occurrence at the injury site and guiding tendon regeneration. Thus, this review is aimed at giving an overview on the role exerted from different tissue engineering actors in leading immunoregeneration by crosstalking with stem and immune cells to generate new paradigms in designing regenerative medicine approaches for tendon injuries.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
- Correspondence:
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Liliana Liverani
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Aldo R. Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
49
|
Brannon ER, Guevara MV, Pacifici NJ, Lee JK, Lewis JS, Eniola-Adefeso O. Polymeric particle-based therapies for acute inflammatory diseases. NATURE REVIEWS. MATERIALS 2022; 7:796-813. [PMID: 35874960 PMCID: PMC9295115 DOI: 10.1038/s41578-022-00458-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 05/02/2023]
Abstract
Acute inflammation is essential for initiating and coordinating the body's response to injuries and infections. However, in acute inflammatory diseases, inflammation is not resolved but propagates further, which can ultimately lead to tissue damage such as in sepsis, acute respiratory distress syndrome and deep vein thrombosis. Currently, clinical protocols are limited to systemic steroidal treatments, fluids and antibiotics that focus on eradicating inflammation rather than modulating it. Strategies based on stem cell therapeutics and selective blocking of inflammatory molecules, despite showing great promise, still lack the scalability and specificity required to treat acute inflammation. By contrast, polymeric particle systems benefit from uniform manufacturing at large scales while preserving biocompatibility and versatility, thus providing an ideal platform for immune modulation. Here, we outline design aspects of polymeric particles including material, size, shape, deformability and surface modifications, providing a strategy for optimizing the targeting of acute inflammation.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | | | - Noah J. Pacifici
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | - Jonathan K. Lee
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | | |
Collapse
|
50
|
Yu Y, Xing L, Li L, Wu J, He J, Huang Y. Coordination of rigidity modulation and targeting ligand modification on orally-delivered nanoparticles for the treatment of liver fibrosis. J Control Release 2021; 341:215-226. [PMID: 34822908 DOI: 10.1016/j.jconrel.2021.11.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 02/05/2023]
Abstract
Although the individual role of ligand modification or rigidity modulation on oral administration of nanoparticle (NP) has been investigated, how they mutually affect each other remains to be elucidated. Here, we fabricated different rigidity NP with or without surface decoration of FcBP, a neonatal Fc receptor domain-binding peptide. In vitro studies showed that, without FcBP modification, stiff NP had higher transcytosis efficiency across the epithelium than softer NP, due to the different endocytosis mechanisms, intracellular trafficking routes, and exocytosis rate. Notably, after FcBP modification, such difference was narrowed, in a manner that was more favorable for softer NP to "catch up" with stiff NP, suggesting ligand modification was more conducive to exert transcytosis-promoting efficacy on softer NP. In vivo experiments demonstrated that, for ligand-free NP, high rigidity was required for efficient oral absorption and liver distribution. Further FcBP modification decreased that "rigidity threshold", and expanded the feasible rigidity range from stiff NP to softer NP. Upon oral administration, FcBP-modified dexamethasone-loaded softer NP achieved a therapeutic efficacy comparable with stiff NP on alleviating liver fibrosis. Collectively, our study highlighted the necessity of coordinating ligand modification and rigidity modulation for oral drug delivery.
Collapse
Affiliation(s)
- Yinglan Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - LiYun Xing
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiawei Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|