1
|
Mathur R, Elsafy S, Press AT, Brück J, Hornef M, Martin L, Schürholz T, Marx G, Bartneck M, Kiessling F, Metselaar JM, Storm G, Lammers T, Sofias AM, Koczera P. Neutrophil Hitchhiking Enhances Liposomal Dexamethasone Therapy of Sepsis. ACS NANO 2024; 18:28866-28880. [PMID: 39393087 DOI: 10.1021/acsnano.4c09054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Sepsis is characterized by a dysregulated immune response and is very difficult to treat. In the cecal ligation and puncture (CLP) mouse model, we show that nanomedicines can effectively alleviate systemic and local septic events by targeting neutrophils. Specifically, by decorating the surface of clinical-stage dexamethasone liposomes with cyclic arginine-glycine-aspartic acid (cRGD) peptides, we promote their engagement with neutrophils in the systemic circulation, leading to their prominent accumulation at primary and secondary sepsis sites. cRGD-targeted dexamethasone liposomes potently reduce immature circulating neutrophils and neutrophil extracellular traps in intestinal sepsis induction sites and the liver. Additionally, they mitigate inflammatory cytokines systemically and locally while preserving systemic IL-10 levels, contributing to lower IFN-γ/IL-10 ratios as compared to control liposomes and free dexamethasone. Our strategy addresses sepsis at the cellular level, illustrating the use of neutrophils both as a therapeutic target and as a chariot for drug delivery.
Collapse
Affiliation(s)
- Ritvik Mathur
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Sara Elsafy
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
- Medical Faculty, Friedrich-Schiller-University, Jena 07747, Germany
| | - Julian Brück
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Lukas Martin
- Department of Intensive and Intermediate Care Medicine, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Tobias Schürholz
- Department of Intensive and Intermediate Care Medicine, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Gernot Marx
- Department of Intensive and Intermediate Care Medicine, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Matthias Bartneck
- Department of Medicine III, Medical Faculty, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Josbert Maarten Metselaar
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Gert Storm
- Department of Pharmaceutics, Faculty of Science, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
- Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
- Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Patrick Koczera
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen 52074, Germany
- Department of Intensive and Intermediate Care Medicine, RWTH Aachen University Hospital, Aachen 52074, Germany
| |
Collapse
|
2
|
Li C, Liu Q, Han L, Zhang H, Immler R, Rathkolb B, Secklehner J, de Angelis MH, Yildirim AÖ, Zeuschner D, Nicke A, Carlin LM, Sperandio M, Stoeger T, Rehberg M. The eATP/P2×7R Axis Drives Quantum Dot-Nanoparticle Induced Neutrophil Recruitment in the Pulmonary Microcirculation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2404661. [PMID: 39364760 DOI: 10.1002/advs.202404661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/30/2024] [Indexed: 10/05/2024]
Abstract
Exposure to nanoparticles (NPs) is frequently associated with adverse cardiovascular effects. In contrast, NPs in nanomedicine hold great promise for precise lung-specific drug delivery, especially considering the extensive pulmonary capillary network that facilitates interactions with bloodstream-suspended particles. Therefore, exact knowledge about effects of engineered NPs within the pulmonary microcirculation are instrumental for future application of this technology in patients. To unravel the real-time dynamics of intravenously delivered NPs and their effects in the pulmonary microvasculature, we employed intravital microscopy of the mouse lung. Only PEG-amine-QDs, but not carboxyl-QDs triggered rapid neutrophil recruitment in microvessels and their subsequent recruitment to the alveolar space and was linked to cellular degranulation, TNF-α, and DAMP release into the circulation, particularly eATP. Stimulation of the ATP-gated receptor P2X7R induced expression of E-selectin on microvascular endothelium thereby mediating the neutrophilic immune response. Leukocyte integrins LFA-1 and MAC-1 facilitated adhesion and decelerated neutrophil crawling on the vascular surface. In summary, this study unravels the complex cascade of neutrophil recruitment during NP-induced sterile inflammation. Thereby we demonstrate novel adverse effects for NPs in the pulmonary microcirculation and provide critical insights for optimizing NP-based drug delivery and therapeutic intervention strategies, to ensure their efficacy and safety in clinical applications.
Collapse
Affiliation(s)
- Chenxi Li
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
- Department of Pulmonary and Critical Care, Shandong Provincial Hospital Affiliated to, Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiongliang Liu
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Lianyong Han
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Haiyun Zhang
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Roland Immler
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152, Planegg-Martinsried, Germany
| | - Birgit Rathkolb
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Experimental Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Judith Secklehner
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, 85354, Freising, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
- Institute of Experimental Pneumology, LMU, 80539, Munich, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, 48149, Muenster, Germany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 80336, Munich, Germany
| | - Leo M Carlin
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152, Planegg-Martinsried, Germany
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Markus Rehberg
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| |
Collapse
|
3
|
Kim C, Kim H, Sim WS, Jung M, Hong J, Moon S, Park JH, Kim JJ, Kang M, Kwon S, Kim MJ, Ban K, Park HJ, Kim BS. Spatiotemporal control of neutrophil fate to tune inflammation and repair for myocardial infarction therapy. Nat Commun 2024; 15:8481. [PMID: 39353987 PMCID: PMC11445496 DOI: 10.1038/s41467-024-52812-6] [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: 02/15/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Neutrophils are critical mediators of both the initiation and resolution of inflammation after myocardial infarction (MI). Overexuberant neutrophil signaling after MI exacerbates cardiomyocyte apoptosis and cardiac remodeling while neutrophil apoptosis at the injury site promotes macrophage polarization toward a pro-resolving phenotype. Here, we describe a nanoparticle that provides spatiotemporal control over neutrophil fate to both stymie MI pathogenesis and promote healing. Intravenous injection of roscovitine/catalase-loaded poly(lactic-co-glycolic acid) nanoparticles after MI leads to nanoparticle uptake by circulating neutrophils migrating to the infarcted heart. Activated neutrophils at the infarcted heart generate reactive oxygen species, triggering intracellular release of roscovitine, a cyclin-dependent kinase inhibitor, from the nanoparticles, thereby inducing neutrophil apoptosis. Timely apoptosis of activated neutrophils at the infarcted heart limits neutrophil-driven inflammation, promotes macrophage polarization toward a pro-resolving phenotype, and preserves heart function. Modulating neutrophil fate to tune both inflammatory and reparatory processes may be an effective strategy to treat MI.
Collapse
Affiliation(s)
- Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Mikyung Kang
- School of Health and Environmental Science, Korea University, Seoul, Republic of Korea
| | - Sungpil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Mi-Jeong Kim
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea.
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea.
- Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.
- Institute of Engineering Research, Seoul National University, Seoul, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Zamora ME, Essien EO, Bhamidipati K, Murthy A, Liu J, Kim H, Patel MN, Nong J, Wang Z, Espy C, Chaudhry FN, Ferguson LT, Tiwari S, Hood ED, Marcos-Contreras OA, Omo-Lamai S, Shuvaeva T, Arguiri E, Wu J, Rauova L, Poncz M, Basil MC, Cantu E, Planer JD, Spiller K, Zepp J, Muzykantov VR, Myerson JW, Brenner JS. Marginated Neutrophils in the Lungs Effectively Compete for Nanoparticles Targeted to the Endothelium, Serving as a Part of the Reticuloendothelial System. ACS NANO 2024; 18:22275-22297. [PMID: 39105696 DOI: 10.1021/acsnano.4c06286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Nanomedicine has long pursued the goal of targeted delivery to specific organs and cell types but has yet to achieve this goal with the vast majority of targets. One rare example of success in this pursuit has been the 25+ years of studies targeting the lung endothelium using nanoparticles conjugated to antibodies against endothelial surface molecules. However, here we show that such "endothelial-targeted" nanocarriers also effectively target the lungs' numerous marginated neutrophils, which reside in the pulmonary capillaries and patrol for pathogens. We show that marginated neutrophils' uptake of many of these "endothelial-targeted" nanocarriers is on par with endothelial uptake. This generalizes across diverse nanomaterials and targeting moieties and was even found with physicochemical lung tropism (i.e., without targeting moieties). Further, we observed this in ex vivo human lungs and in vivo healthy mice, with an increase in marginated neutrophil uptake of nanoparticles caused by local or distant inflammation. These findings have implications for nanomedicine development for lung diseases. These data also suggest that marginated neutrophils, especially in the lungs, should be considered a major part of the reticuloendothelial system (RES), with a special role in clearing nanoparticles that adhere to the lumenal surfaces of blood vessels.
Collapse
Affiliation(s)
- Marco E Zamora
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Eno-Obong Essien
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Aditi Murthy
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Jing Liu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Hyunjun Kim
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Manthan N Patel
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jia Nong
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Carolann Espy
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Fatima N Chaudhry
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Laura T Ferguson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Sachchidanand Tiwari
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Evguenia Arguiri
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jichuan Wu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Lubica Rauova
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Mortimer Poncz
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Maria C Basil
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Edward Cantu
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Joseph D Planer
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kara Spiller
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
| | - Jarod Zepp
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
5
|
Kupor D, Felder ML, Kodikalla S, Chu X, Eniola-Adefeso O. Nanoparticle-neutrophils interactions for autoimmune regulation. Adv Drug Deliv Rev 2024; 209:115316. [PMID: 38663550 PMCID: PMC11246615 DOI: 10.1016/j.addr.2024.115316] [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: 10/21/2023] [Revised: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
Neutrophils play an essential role as 'first responders' in the immune response, necessitating many immune-modulating capabilities. Chronic, unresolved inflammation is heavily implicated in the progression and tissue-degrading effects of autoimmune disease. Neutrophils modulate disease pathogenesis by interacting with the inflammatory and autoreactive cells through effector functions, including signaling, degranulation, and neutrophil extracellular traps (NETs) release. Since the current gold standard systemic glucocorticoid administration has many drawbacks and side effects, targeting neutrophils in autoimmunity provides a new approach to developing therapeutics. Nanoparticles enable targeting of specific cell types and controlled release of a loaded drug cargo. Thus, leveraging nanoparticle properties and interactions with neutrophils provides an exciting new direction toward novel therapies for autoimmune diseases. Additionally, recent work has utilized neutrophil properties to design novel targeted particles for delivery into previously inaccessible areas. Here, we outline nanoparticle-based strategies to modulate neutrophil activity in autoimmunity, including various nanoparticle formulations and neutrophil-derived targeting.
Collapse
Affiliation(s)
- Daniel Kupor
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael L Felder
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shivanie Kodikalla
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xueqi Chu
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Liu X, Ou X, Zhang T, Li X, Qiao Q, Jia L, Xu Z, Zhang F, Tian T, Lan H, Yang C, Kong L, Zhang Z. In situ neutrophil apoptosis and macrophage efferocytosis mediated by Glycyrrhiza protein nanoparticles for acute inflammation therapy. J Control Release 2024; 369:215-230. [PMID: 38508529 DOI: 10.1016/j.jconrel.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
In the progression of acute inflammation, the activation and recruitment of macrophages and neutrophils are mutually reinforcing, leading to amplified inflammatory response and severe tissue damage. Therefore, to regulate the axis of neutrophils and macrophages is essential to avoid tissue damage induced from acute inflammatory. Apoptotic neutrophils can regulate the anti-inflammatory activity of macrophages through the efferocytosis. The strategy of in situ targeting and inducing neutrophil apoptosis has the potential to modulate macrophage activity and transfer anti-inflammatory drugs. Herein, a natural glycyrrhiza protein nanoparticle loaded with dexamethasone (Dex@GNPs) was constructed, which could simultaneously regulate neutrophil and macrophage function during acute inflammation treatment by combining in situ neutrophil apoptosis and macrophage efferocytosis. Dex@GNPs can be rapidly and selectively internalized by neutrophils and subsequently induce neutrophils apoptosis through a ROS-dependent mechanism. The efferocytosis of apoptotic neutrophils not only promoted the polarization of macrophages into anti-inflammatory state, but also facilitated the transfer of Dex@GNPs to macrophages. This enabled dexamethasone to further modulate macrophage function. In mouse models of acute respiratory distress syndrome and sepsis, Dex@GNPs significantly ameliorated the disordered immune microenvironment and alleviated tissue injury. This study presents a novel strategy for drug delivery and inflammation regulation to effectively treat acute inflammatory diseases.
Collapse
Affiliation(s)
- Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangjun Ou
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiantian Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaonan Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Qiao
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liyuan Jia
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhangxi Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fangming Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyi Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Li Y, Moein Moghimi S, Simberg D. Complement-dependent uptake of nanoparticles by blood phagocytes: brief overview and perspective. Curr Opin Biotechnol 2024; 85:103044. [PMID: 38091875 PMCID: PMC11214757 DOI: 10.1016/j.copbio.2023.103044] [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: 11/07/2023] [Accepted: 11/22/2023] [Indexed: 02/09/2024]
Abstract
Immune recognition and uptake of nanoparticles remain the hot topic in nanomedicine research. Complement is the central player in the immune recognition of engineered nanoparticles. Here, we summarize the accumulated knowledge on the role of complement in the interactions of nanomaterials with blood phagocytes. We describe the interplay between surface properties, complement opsonization, and immune uptake, primarily of iron oxide nanoparticles. We discuss the rigor of the published research and further identify the following knowledge gaps: 1) the role of complement in the variability of uptake of nanomaterials in healthy and diseased subjects, and 2) modulation of complement interactions to improve the performance of nanomaterials. Addressing these gaps is critical to improving translational chances of nanomaterials for drug delivery and imaging applications.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Seyed Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, USA; Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
8
|
Emilsson G, Liu K, Höök F, Svensson L, Rosengren L, Lindfors L, Sigfridsson K. The In Vivo Fate of Polycatecholamine Coated Nanoparticles Is Determined by a Fibrinogen Enriched Protein Corona. ACS NANO 2023; 17:24725-24742. [PMID: 38088920 DOI: 10.1021/acsnano.3c04968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Polycatecholamine coatings have attracted significant attention in the past 10 years owing to their ability to functionalize a wide range of materials. Here we apply the use of such coatings to drug nanocrystals, made from a poorly soluble drug compound, to postfunctionalize the nanocrystal surface with the aim of providing steric stabilization and extending their circulation time after intravenous injection. We show that both polydopamine and polynorepinephrine can be used to successfully modify drug nanocrystals and subsequently incorporate end-functionalized PEG to the surface. Even though high grafting densities of PEG were achieved, we observed rapid clearance and increased liver uptake for polycatecholamine functionalized drug nanocrystals. Using both surface sensitive model systems and protein corona profiling, we determine that the rapid clearance was correlated with an increase in adsorption of proteins involved in coagulation to the polycatecholamine surface, with fibrinogen being the most abundant. Further analysis of the most abundant proteins revealed a significant increase in thiol-rich proteins on polycatecholamine coated surfaces. The observed interaction with coagulation proteins highlights one of the current challenges using polycatecholamines for drug delivery but might also provide insights to the growing use of these materials in hemostatic applications.
Collapse
Affiliation(s)
- Gustav Emilsson
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Kai Liu
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, 41296 Gothenburg, Sweden
| | - Lena Svensson
- Bioscience Renal In Vivo Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Louise Rosengren
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Lennart Lindfors
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Kalle Sigfridsson
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| |
Collapse
|
9
|
Xu S, Xu C, Xu J, Zhang K, Zhang H. Macrophage Heterogeneity and Its Impact on Myocardial Ischemia-Reperfusion Injury: An Integrative Review. J Inflamm Res 2023; 16:5971-5987. [PMID: 38088942 PMCID: PMC10712254 DOI: 10.2147/jir.s436560] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/30/2023] [Indexed: 10/21/2024] Open
Abstract
The coronary reperfusion following acute myocardial infarction can paradoxically trigger myocardial ischemia-reperfusion (IR) injury. This complex phenomenon involves the intricate interplay of different subsets of macrophages. These macrophages are crucial players in the post-infarction inflammatory response and subsequent myocardial anti-inflammatory repair. However, their diverse functions can lead to both beneficial and detrimental effects. On one hand, these macrophages play a crucial role in orchestrating the inflammatory response, aiding in the clearance of cellular debris and initiating tissue repair mechanisms. On the other hand, their excessive infiltration and activation can contribute to the perpetuation of the inflammatory cascade, leading to additional myocardial injury and adverse cardiac remodeling. Multiple mechanisms contribute to the IR injury mediated by macrophages, including oxidative stress, apoptosis, and autophagy. These processes further exacerbate the damage to the already vulnerable myocardial tissue. To address this delicate balance, therapeutic strategies aiming to target and modulate macrophage polarization and function are being explored. By fine-tuning the immune inflammatory response, such interventions hold promise in mitigating post-infarction myocardial injury and fostering a more favorable environment for myocardial healing and recovery. Through advancements in this area of research, potential anti-inflammatory interventions may pave the way for improved clinical outcomes and better management of patients after acute myocardial infarction.
Collapse
Affiliation(s)
- Shuwan Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Cong Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Jiahua Xu
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| | - Kun Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Huanji Zhang
- Cardiovascular Department, the Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
10
|
Garanina AS, Vishnevskiy DA, Chernysheva AA, Valikhov MP, Malinovskaya JA, Lazareva PA, Semkina AS, Abakumov MA, Naumenko VA. Neutrophil as a Carrier for Cancer Nanotherapeutics: A Comparative Study of Liposome, PLGA, and Magnetic Nanoparticles Delivery to Tumors. Pharmaceuticals (Basel) 2023; 16:1564. [PMID: 38004431 PMCID: PMC10674452 DOI: 10.3390/ph16111564] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Insufficient drug accumulation in tumors is still a major concern for using cancer nanotherapeutics. Here, the neutrophil-based delivery of three nanoparticle types-liposomes, PLGA, and magnetite nanoparticles-was assessed both in vitro and in vivo. Confocal microscopy and a flow cytometry analysis demonstrated that all the studied nanoparticles interacted with neutrophils from the peripheral blood of mice with 4T1 mammary adenocarcinoma without a significant impact on neutrophil viability or activation state. Intravital microscopy of the tumor microenvironment showed that the neutrophils did not engulf the liposomes after intravenous administration, but facilitated nanoparticle extravasation in tumors through micro- and macroleakages. PLGA accumulated along the vessel walls in the form of local clusters. Later, PLGA nanoparticle-loaded neutrophils were found to cross the vascular barrier and migrate towards the tumor core. The magnetite nanoparticles extravasated in tumors both via spontaneous macroleakages and on neutrophils. Overall, the specific type of nanoparticles largely determined their behavior in blood vessels and their neutrophil-mediated delivery to the tumor. Since neutrophils are the first to migrate to the site of inflammation, they can increase nanodrug delivery effectiveness for nanomedicine application.
Collapse
Affiliation(s)
- Anastasiia S. Garanina
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology «MISIS», 119049 Moscow, Russia;
| | - Daniil A. Vishnevskiy
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.V.); (M.P.V.); (P.A.L.); (A.S.S.)
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia; (A.A.C.); (V.A.N.)
| | - Anastasia A. Chernysheva
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia; (A.A.C.); (V.A.N.)
| | - Marat P. Valikhov
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.V.); (M.P.V.); (P.A.L.); (A.S.S.)
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia; (A.A.C.); (V.A.N.)
| | | | - Polina A. Lazareva
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.V.); (M.P.V.); (P.A.L.); (A.S.S.)
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.V.); (M.P.V.); (P.A.L.); (A.S.S.)
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia; (A.A.C.); (V.A.N.)
| | - Maxim A. Abakumov
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology «MISIS», 119049 Moscow, Russia;
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.V.); (M.P.V.); (P.A.L.); (A.S.S.)
| | - Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, 119034 Moscow, Russia; (A.A.C.); (V.A.N.)
| |
Collapse
|
11
|
Lin P, Gao R, Fang Z, Yang W, Tang Z, Wang Q, Wu Y, Fang J, Yu W. Precise nanodrug delivery systems with cell-specific targeting for ALI/ARDS treatment. Int J Pharm 2023; 644:123321. [PMID: 37591476 DOI: 10.1016/j.ijpharm.2023.123321] [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: 05/18/2023] [Revised: 07/22/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common acute and critical diseases in clinics and have no effective treatment to date. With the concept of "precision medicine", research into the precise drug delivery of therapeutic and diagnostic drugs has become a frontier in nanomedicine research and has entered the era of design of precise nanodrug delivery systems (NDDSs) with cell-specific targeting. Owing to the distinctive characteristics of ALI/ARDS, designing NDDSs for specific focal sites is an important strategy for changing drug distribution in the body and specifically increasing drug concentration at target sites while decreasing drug concentration at non-target sites. This strategy enhances drug efficacy, reduces adverse reactions, and ensures accurate nano-targeted treatment. On the basis of the characteristics of pathological ALI/ARDS microenvironments, this paper reviews NDDSs targeting vascular endothelial cells, neutrophils, alveolar macrophages, and alveolar epithelial cells to provide reference for designing accurate NDDSs for ALI/ARDS and novel insights into targeted treatments for ALI/ARDS.
Collapse
Affiliation(s)
- Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Zhan Tang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Qiao Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Yueguo Wu
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Jie Fang
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou 310013, China.
| | - Wenying Yu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
12
|
Banka AL, Guevara MV, Brannon ER, Nguyen NQ, Song S, Cady G, Pinsky DJ, Uhrich KE, Adili R, Holinstat M, Eniola-Adefeso O. Cargo-free particles divert neutrophil-platelet aggregates to reduce thromboinflammation. Nat Commun 2023; 14:2462. [PMID: 37117163 PMCID: PMC10144907 DOI: 10.1038/s41467-023-37990-z] [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: 09/10/2022] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
The combination of inflammation and thrombosis is a hallmark of many cardiovascular diseases. Under such conditions, platelets are recruited to an area of inflammation by forming platelet-leukocyte aggregates via interaction of PSGL-1 on leukocytes and P-selectin on activated platelets, which can bind to the endothelium. While particulate drug carriers have been utilized to passively redirect leukocytes from areas of inflammation, the downstream impact of these carriers on platelet accumulation in thromboinflammatory conditions has yet to be studied. Here, we explore the ability of polymeric particles to divert platelets away from inflamed blood vessels both in vitro and in vivo. We find that untargeted and targeted micron-sized polymeric particles can successfully reduce platelet adhesion to an inflamed endothelial monolayer in vitro in blood flow systems and in vivo in a lipopolysaccharide-induced, systemic inflammation murine model. Our data represent initial work in developing cargo-free, anti-platelet therapeutics specifically for conditions of thromboinflammation.
Collapse
Affiliation(s)
- Alison L Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - M Valentina Guevara
- 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
| | - Nhien Q Nguyen
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Shuang Song
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Gillian Cady
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David J Pinsky
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kathryn E Uhrich
- Department of Chemistry, University of California Riverside, Riverside, CA, 92521, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Holinstat
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Pondman K, Le Gac S, Kishore U. Nanoparticle-induced immune response: Health risk versus treatment opportunity? Immunobiology 2023; 228:152317. [PMID: 36592542 DOI: 10.1016/j.imbio.2022.152317] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Nanoparticles (NPs) are not only employed in many biomedical applications in an engineered form, but also occur in our environment, in a more hazardous form. NPs interact with the immune system through various pathways and can lead to a myriad of different scenarios, ranging from their quiet removal from circulation by macrophages without any impact for the body, to systemic inflammatory effects and immuno-toxicity. In the latter case, the function of the immune system is affected by the presence of NPs. This review describes, how both the innate and adaptive immune system are involved in interactions with NPs, together with the models used to analyse these interactions. These models vary between simple 2D in vitro models, to in vivo animal models, and also include complex all human organ on chip models which are able to recapitulate more accurately the interaction in the in vivo situation. Thereafter, commonly encountered NPs in both the environment and in biomedical applications and their possible effects on the immune system are discussed in more detail. Not all effects of NPs on the immune system are detrimental; in the final section, we review several promising strategies in which the immune response towards NPs can be exploited to suit specific applications such as vaccination and cancer immunotherapy.
Collapse
Affiliation(s)
- Kirsten Pondman
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, University of Twente, Enschede, the Netherlands.
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, University of Twente, Enschede, the Netherlands
| | - Uday Kishore
- Biosciences, Brunel University London, Uxbridge, UK; Department of Veterinary Medicine, U.A.E. University, Al Ain, United Arab Emirates
| |
Collapse
|
14
|
Hui M, Jia X, Li X, Lazcano-Silveira R, Shi M. Anti-Inflammatory and Antioxidant Effects of Liposoluble C60 at the Cellular, Molecular, and Whole-Animal Levels. J Inflamm Res 2023; 16:83-93. [PMID: 36643955 PMCID: PMC9833127 DOI: 10.2147/jir.s386381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/16/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Liposoluble carbon-60 (C60) has potential applications in many fields, including cosmetics, medical devices, and medicine, but its specific mechanism of action remains unclear. This study explored whether liposoluble C60 could be delivered to human organs, tissues, and cells through blood, extracellular fluid, and cell culture fluid and whether it exerts anti-inflammatory and antioxidant effects at the molecular, cellular, and whole-animal levels. Methods At the cellular level, we mixed C60 dissolved in grape seed oil with cell culture medium containing 10% serum and investigated its effects on tumor necrosis factor-α (TNF-α) release, migration, phagocytosis, respiratory burst, and apoptosis in freshly isolated human neutrophils. At the molecular level, we mixed a trace amount of C60 dissolved in grape seed oil with aqueous and ethanolic solutions and studied its antioxidant effect. At the animal level, we investigated the inhibitory effect of C60 on the serum inflammatory marker C-reactive protein (CRP) in beagle dogs after oral administration of C60 dissolved in grape seed oil. Results The results showed that the trace amount of C60 dissolved in grape seed oil significantly inhibited TNF-α release, cell migration, phagocytosis, and respiratory burst in freshly isolated human neutrophils. In addition, the trace amount of C60 dissolved in grape seed oil had a significant scavenging effect on superoxide free radicals and 1,1-diphenyl-2-trinitrophenylhydrazine free radicals. Oral administration of C60 dissolved in grape seed oil markedly reduced the level of the serum inflammatory marker CRP in beagle dogs. Conclusion In summary, a trace amount of hydrophobic C60 in hydrophilic media effectively produced anti-inflammatory and antioxidant effects in cells and animals. C60 dissolved in grape seed oil is a novel anti-inflammatory and antioxidant drug candidate.
Collapse
Affiliation(s)
- Mizhou Hui
- College of Life Sciences, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| | - Xiaoxiao Jia
- College of Life Sciences, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| | - Xinrong Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong, People’s Republic of China
| | - Rayko Lazcano-Silveira
- College of Life Sciences, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| | - Ming Shi
- Harbin Institute of Technology, Harbin, Heilongjiang, People’s Republic of China,Correspondence: Ming Shi, Harbin Institute of Technology, 2 Yikuang Street, Nangang District, Harbin, Heilongjiang, 150001, People’s Republic of China, Tel +86 13654537645, Email
| |
Collapse
|
15
|
Kelley WJ, Wragg KM, Chen J, Murthy T, Xu Q, Boyne MT, Podojil JR, Elhofy A, Goldstein DR. Nanoparticles reduce monocytes within the lungs to improve outcomes after influenza virus infection in aged mice. JCI Insight 2022; 7:156320. [PMID: 35737459 PMCID: PMC9462478 DOI: 10.1172/jci.insight.156320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 06/21/2022] [Indexed: 01/26/2023] Open
Abstract
Older people exhibit dysregulated innate immunity to respiratory viral infections, including influenza and SARS-CoV-2, and show an increase in morbidity and mortality. Nanoparticles are a potential practical therapeutic that could reduce exaggerated innate immune responses within the lungs during viral infection. However, such therapeutics have not been examined for effectiveness during respiratory viral infection, particular in aged hosts. Here, we employed a lethal model of influenza viral infection in vulnerable aged mice to examine the ability of biodegradable, cargo-free nanoparticles, designated ONP-302, to resolve innate immune dysfunction and improve outcomes during infection. We administered ONP-302 via i.v. injection to aged mice at day 3 after infection, when the hyperinflammatory innate immune response was already established. During infection, we found that ONP-302 treatment reduced the numbers of inflammatory monocytes within the lungs and increased their number in both the liver and spleen, without impacting viral clearance. Importantly, cargo-free nanoparticles reduced lung damage, reduced histological lung inflammation, and improved gas exchange and, ultimately, the clinical outcomes in influenza-infected aged mice. In conclusion, ONP-302 improves outcomes in influenza-infected aged mice. Thus, our study provides information concerning a practical therapeutic, which, if translated clinically, could improve disease outcomes for vulnerable older patients suffering from respiratory viral infections.
Collapse
Affiliation(s)
| | | | - Judy Chen
- Department of Internal Medicine and,Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tushar Murthy
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Qichen Xu
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Michael T. Boyne
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Joseph R. Podojil
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Adam Elhofy
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine and,Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Microbiology and Immunology, University of Michigan, Michigan, USA
| |
Collapse
|
16
|
Maisha N, Kulkarni C, Pandala N, Zilberberg R, Schaub L, Neidert L, Glaser J, Cannon J, Janeja V, Lavik EB. PEGylated Polyester Nanoparticles Trigger Adverse Events in a Large Animal Model of Trauma and in Naı̈ve Animals: Understanding Cytokine and Cellular Correlations with These Events. ACS NANO 2022; 16:10566-10580. [PMID: 35822898 DOI: 10.1021/acsnano.2c01993] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Intravenously infusible nanoparticles to control bleeding have shown promise in rodents, but translation into preclinical models has been challenging as many of these nanoparticle approaches have resulted in infusion responses and adverse outcomes in large animal trauma models. We developed a hemostatic nanoparticle technology that was screened to avoid one component of the infusion response: complement activation. We administered these hemostatic nanoparticles, control nanoparticles, or saline volume controls in a porcine polytrauma model. While the hemostatic nanoparticles promoted clotting as marked by a decrease in prothrombin time and both the hemostatic nanoparticles and controls did not active complement, in a subset of the animals, hard thrombi were found in uninjured tissues in both the hemostatic and control nanoparticle groups. Using data science methods that allow one to work across heterogeneous data sets, we found that the presence of these thrombi correlated with changes in IL-6, INF-alpha, lymphocytes, and neutrophils. While these findings might suggest that this formulation would not be a safe one for translation for trauma, they provide guidance for developing screening tools to make nanoparticle formulations in the complex milieux of trauma as well as for therapeutic interventions more broadly. This is important as we look to translate intravenously administered nanoparticle formulations for therapies, particularly considering the vascular changes seen in a subset of patients following COVID-19. We need to understand adverse events like thrombi more completely and screen for these events early to make nanomaterials as safe and effective as possible.
Collapse
Affiliation(s)
| | | | | | | | - Leasha Schaub
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Leslie Neidert
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Jacob Glaser
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Jeremy Cannon
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | | | | |
Collapse
|
17
|
Moreno-Mendieta S, Guillén D, Vasquez-Martínez N, Hernández-Pando R, Sánchez S, Rodríguez-Sanoja R. Understanding the Phagocytosis of Particles: the Key for Rational Design of Vaccines and Therapeutics. Pharm Res 2022; 39:1823-1849. [PMID: 35739369 DOI: 10.1007/s11095-022-03301-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/23/2022] [Indexed: 12/17/2022]
Abstract
A robust comprehension of phagocytosis is crucial for understanding its importance in innate immunity. A detailed description of the molecular mechanisms that lead to the uptake and clearance of endogenous and exogenous particles has helped elucidate the role of phagocytosis in health and infectious or autoimmune diseases. Furthermore, knowledge about this cellular process is important for the rational design and development of particulate systems for the administration of vaccines or therapeutics. Depending on these specific applications and the required biological responses, particles must be designed to encourage or avoid their phagocytosis and prolong their circulation time. Functionalization with specific polymers or ligands and changes in the size, shape, or surface of particles have important effects on their recognition and internalization by professional and nonprofessional phagocytes and have a major influence on their fate and safety. Here, we review the phagocytosis of particles intended to be used as carrier or delivery systems for vaccines or therapeutics, the cells involved in this process depending on the route of administration, and the strategies employed to obtain the most desirable particles for each application through the manipulation of their physicochemical characteristics. We also offer a view of the challenges and potential opportunities in the field and give some recommendations that we expect will enable the development of improved approaches for the rational design of these systems.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México, Mexico. .,Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| | - Daniel Guillén
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Nathaly Vasquez-Martínez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.,Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Romina Rodríguez-Sanoja
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| |
Collapse
|
18
|
Li H, Yang YG, Sun T. Nanoparticle-Based Drug Delivery Systems for Induction of Tolerance and Treatment of Autoimmune Diseases. Front Bioeng Biotechnol 2022; 10:889291. [PMID: 35464732 PMCID: PMC9019755 DOI: 10.3389/fbioe.2022.889291] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune disease is a chronic inflammatory disease caused by disorders of immune regulation. Antigen-specific immunotherapy has the potential to inhibit the autoreactivity of inflammatory T cells and induce antigen-specific immune suppression without impairing normal immune function, offering an ideal strategy for autoimmune disease treatment. Tolerogenic dendritic cells (Tol DCs) with immunoregulatory functions play important roles in inducing immune tolerance. However, the effective generation of tolerogenic DCs in vivo remains a great challenge. The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance in vivo. In this review, we discuss multiple nanoparticles, with a focus on their potential in treatment of autoimmune diseases. We also discuss how the physical properties of nanoparticles influence their therapeutic efficacy.
Collapse
Affiliation(s)
- He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- Department of Rehabilitation Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| |
Collapse
|
19
|
Brannon ER, Kelley WJ, Newstead MW, Banka AL, Uhrich KE, O’Connor CE, Standiford TJ, Eniola-Adefeso O. Polysalicylic Acid Polymer Microparticle Decoys Therapeutically Treat Acute Respiratory Distress Syndrome. Adv Healthc Mater 2022; 11:e2101534. [PMID: 34881524 PMCID: PMC8986552 DOI: 10.1002/adhm.202101534] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/03/2021] [Indexed: 01/13/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) remain problematic due to high mortality rates and lack of effective treatments. Neutrophilic injury contributes to mortality in ALI/ARDS. Here, technology for rapid ARDS intervention is developed and evaluated, where intravenous salicylic acid-based polymer microparticles, i.e., Poly-Aspirin (Poly-A), interfere with neutrophils in blood, reducing lung neutrophil infiltration and injury in vivo in mouse models of ALI/ARDS. Importantly, Poly-A particles reduce multiple inflammatory cytokines in the airway and bacterial load in the bloodstream in a live bacteria lung infection model of ARDS, drastically improving survival. It is observed that phagocytosis of the Poly-A microparticles, with salicylic acid in the polymer backbone, alters the neutrophil surface expression of adhesion molecules, potentially contributing to their added therapeutic benefits. Given the proven safety profile of the microparticle degradation products-salicylic acid and adipic acid-it is anticipated that the Poly-A particles represent a therapeutic strategy in ARDS with a rare opportunity for rapid clinical translation.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - William J. Kelley
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | | | - Alison L. Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Kathryn E. Uhrich
- Department of Chemistry, University of California Riverside, Riverside, CA
| | | | | | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
20
|
Liu J, Liu Z, Pang Y, Zhou H. The interaction between nanoparticles and immune system: application in the treatment of inflammatory diseases. J Nanobiotechnology 2022; 20:127. [PMID: 35279135 PMCID: PMC8917374 DOI: 10.1186/s12951-022-01343-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Nanoparticle (NP) is an emerging tool applied in the biomedical field. With combination of different materials and adjustment of their physical and chemical properties, nanoparticles can have diverse effects on the organism and may change the treating paradigm of multiple diseases in the future. More and more results show that nanoparticles can function as immunomodulators and some formulas have been approved for the treatment of inflammation-related diseases. However, our current understanding of the mechanisms that nanoparticles can influence immune responses is still limited, and systemic clinical trials are necessary for the evaluation of their security and long-term effects. This review provides an overview of the recent advances in nanoparticles that can interact with different cellular and molecular components of the immune system and their application in the management of inflammatory diseases, which are caused by abnormal immune reactions. This article focuses on the mechanisms of interaction between nanoparticles and the immune system and tries to provide a reference for the future design of nanotechnology for the treatment of inflammatory diseases.
Collapse
|
21
|
Sharma P, Vijaykumar A, Raghavan JV, Rananaware SR, Alakesh A, Bodele J, Rehman JU, Shukla S, Wagde V, Nadig S, Chakrabarti S, Visweswariah SS, Nandi D, Gopal B, Jhunjhunwala S. Particle uptake driven phagocytosis in macrophages and neutrophils enhances bacterial clearance. J Control Release 2022; 343:131-141. [PMID: 35085696 PMCID: PMC7615985 DOI: 10.1016/j.jconrel.2022.01.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022]
Abstract
Humans are exposed to numerous synthetic foreign particles in the form of drug delivery systems and diagnostic agents. Specialized immune cells (phagocytes) clear these particles by phagocytosing and attempting to degrade them. The process of recognition and internalization of the particles may trigger changes in the function of phagocytes. Some of these changes, especially the ability of a particle-loaded phagocyte to take up and neutralize pathogens, remains poorly studied. Herein, we demonstrate that the uptake of non-stimulatory cargo-free particles enhances the phagocytic ability of monocytes, macrophages and neutrophils. The enhancement in phagocytic ability was independent of particle properties, such as size or the base material constituting the particle. Additionally, we show that the increased phagocytosis was not a result of cellular activation or cellular heterogeneity but was driven by changes in cell membrane fluidity and cellular compliance. A consequence of the enhanced phagocytic activity was that particulate-laden immune cells neutralize Escherichia coli (E. coli) faster in culture. Moreover, when administered in mice as a prophylactic, particulates enable faster clearance of E. coli and Staphylococcus epidermidis. Together, we demonstrate that the process of uptake induces cellular changes that favor additional phagocytic events. This study provides insights into using non-stimulatory cargo-free particles to engineer immune cell functions for applications involving faster clearance of phagocytosable abiotic and biotic material.
Collapse
Affiliation(s)
- Preeti Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Anjali Vijaykumar
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | | | | | - Alakesh Alakesh
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Janhavi Bodele
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Junaid Ur Rehman
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Shivani Shukla
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Virta Wagde
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Savitha Nadig
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru 560012, India
| | - Sveta Chakrabarti
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560012, India
| | | | - Siddharth Jhunjhunwala
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India.
| |
Collapse
|
22
|
Snoderly HT, Freshwater KA, Martinez de la Torre C, Panchal DM, Vito JN, Bennewitz MF. PEGylation of Metal Oxide Nanoparticles Modulates Neutrophil Extracellular Trap Formation. BIOSENSORS 2022; 12:123. [PMID: 35200382 PMCID: PMC8869785 DOI: 10.3390/bios12020123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/04/2022] [Accepted: 02/13/2022] [Indexed: 06/01/2023]
Abstract
Novel metal oxide nanoparticle (NP) contrast agents may offer safety and functionality advantages over conventional gadolinium-based contrast agents (GBCAs) for cancer diagnosis by magnetic resonance imaging. However, little is known about the behavior of metal oxide NPs, or of their effect, upon coming into contact with the innate immune system. As neutrophils are the body's first line of defense, we sought to understand how manganese oxide and iron oxide NPs impact leukocyte functionality. Specifically, we evaluated whether contrast agents caused neutrophils to release web-like fibers of DNA known as neutrophil extracellular traps (NETs), which are known to enhance metastasis and thrombosis in cancer patients. Murine neutrophils were treated with GBCA, bare manganese oxide or iron oxide NPs, or poly(lactic-co-glycolic acid) (PLGA)-coated metal oxide NPs with different incorporated levels of poly(ethylene glycol) (PEG). Manganese oxide NPs elicited the highest NETosis rates and had enhanced neutrophil uptake properties compared to iron oxide NPs. Interestingly, NPs with low levels of PEGylation produced more NETs than those with higher PEGylation. Despite generating a low rate of NETosis, GBCA altered neutrophil cytokine expression more than NP treatments. This study is the first to investigate whether manganese oxide NPs and GBCAs modulate NETosis and reveals that contrast agents may have unintended off-target effects which warrant further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | - Margaret F. Bennewitz
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (H.T.S.); (K.A.F.); (C.M.d.l.T.); (D.M.P.); (J.N.V.)
| |
Collapse
|
23
|
Jarai BM, Fromen CA. Nanoparticle Internalization Promotes the Survival of Primary Macrophages. ADVANCED NANOBIOMED RESEARCH 2022; 2. [PMID: 35991157 PMCID: PMC9387674 DOI: 10.1002/anbr.202100127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Macrophages, a class of tissue resident innate immune cells, are responsible for sequestering foreign objects through the process of phagocytosis, making them a promising target for immune-modulation via particulate engineering. Here, we report that nanoparticle (NP) dosing and cellular internalization via phagocytosis significantly enhances survival of ex vivo cultures of primary bone marrow-derived, alveolar, and peritoneal macrophages over particle-free controls. The enhanced survival is attributed to suppression of caspase-dependent apoptosis and is linked to phagocytosis and lysosomal signaling. Uniquely, poly(ethylene glycol)-based NP treatment extended cell viability in the absence of macrophage polarization and enhanced expression of pro-survival B cell lymphoma-2 (Bcl-2) protein in macrophages following multiple routes of in vivo administration. The enhanced survival phenomenon is also applicable to NPs of alternative chemistries, indicating the potential universality of this phenomenon with relevant drug delivery particles. These findings provide a framework for extending the lifespan of primary macrophages ex vivo for drug screening, vaccine studies, and cell therapies and has implications for any in vivo particulate immune-engineering applications.
Collapse
Affiliation(s)
- Bader M. Jarai
- Department of Chemical and Biomolecular Engineering University of Delaware 150 Academy St. Newark DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering University of Delaware 150 Academy St. Newark DE 19716 USA
| |
Collapse
|
24
|
Wang Y, Hou M, Duan S, Zhao Z, Wu X, Chen Y, Yin L. Macrophage-targeting gene silencing orchestrates myocardial microenvironment remodeling toward the anti-inflammatory treatment of ischemia-reperfusion (IR) injury. Bioact Mater 2022; 17:320-333. [PMID: 35386446 PMCID: PMC8965030 DOI: 10.1016/j.bioactmat.2022.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/02/2022] [Accepted: 01/18/2022] [Indexed: 01/03/2023] Open
Abstract
Ischemia-reperfusion (IR) injury represents a major cause of myocardial dysfunction after infarction and thrombolytic therapy, and it is closely related to the free radical explosion and overwhelming inflammatory responses. Herein, macrophage-targeting nanocomplexes (NCs) are developed to mediate efficient co-delivery of siRNA against MOF (siMOF) and microRNA-21 (miR21) into myocardial macrophages, cooperatively orchestrating the myocardial microenvironment against IR injury. Bioreducible, branched poly(β-amino ester) (BPAE-SS) is designed to co-condense siMOF and miR21 into NCs in a multivalency-reinforced approach, and they are surface-decorated with carboxylated mannan (Man-COOH) to shield the positive surface charges and enhance the serum stability. The final MBSsm NCs are efficiently internalized by myocardial macrophages after systemic administration, wherein BPAE-SS is degraded into small segments by intracellular glutathione to promote the siMOF/miR21 release, finally provoking efficient gene silencing. Thus, cardiomyocyte protection and macrophage modulation are realized via the combined effects of ROS scavenging, inflammation inhibition, and autophagy attenuation, which ameliorates the myocardial microenvironment and restores the cardiac function via positive cellular crosstalk. This study renders promising solutions to address the multiple systemic barriers against in vivo nucleic acid delivery, and it also offers new options for IR injury by manipulating multiple reciprocal bio-reactions. Macrophage-targeting and reduction-dissociable NCs mediate efficient siMOF/miR21 co-delivery. siMOF and miR21 cooperatively inhibit ROS production, inflammation, and autophagy. siMOF and miR21 orchestrate microenvironment remodeling via cellular cross-talk. NCs mediate efficient treatment of myocardial ischemia reperfusion injury.
Collapse
|
25
|
The consequences of particle uptake on immune cells. Trends Pharmacol Sci 2022; 43:305-320. [DOI: 10.1016/j.tips.2022.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022]
|
26
|
Shanmugam H, Rengarajan C, Nataraj S, Sharma A. Interactions of plant food bioactives‐loaded nano delivery systems at the nano‐bio interface and its pharmacokinetics: An overview. FOOD FRONTIERS 2022. [DOI: 10.1002/fft2.130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Haripriya Shanmugam
- Department of Nano Science and Technology Tamil Nadu Agricultural University Tamil Nadu India
| | - Chitra Rengarajan
- Department of Nano Science and Technology Tamil Nadu Agricultural University Tamil Nadu India
| | - Swathika Nataraj
- Department of Nano Science and Technology Tamil Nadu Agricultural University Tamil Nadu India
| | - Aashima Sharma
- Department of Chemistry Panjab University Chandigarh India
| |
Collapse
|
27
|
Jiang H, Wang X, Li X, Jin Y, Yan Z, Yao X, Yuan WE, Qian Y, Ouyang Y. A multifunctional ATP-generating system by reduced graphene oxide-based scaffold repairs neuronal injury by improving mitochondrial function and restoring bioelectricity conduction. Mater Today Bio 2022; 13:100211. [PMID: 35198959 PMCID: PMC8841887 DOI: 10.1016/j.mtbio.2022.100211] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 01/09/2023] Open
Abstract
Peripheral nerve injury usually impairs neurological functions. The excessive oxidative stress and disrupted bioelectrical conduction gives rise to a hostile microenvironment and impedes nerve regeneration. Therefore, it is of urgent need to develop tissue engineering products which help alleviate the oxidative insults and restore bioelectrical signals. Melatonin (MLT) is an important endogenous hormone that diminishes the accumulation of reactive oxygen species. Reduced graphene oxide (RGO) possesses the excellent electrical conductivity and biocompatibility. In this study, a multilayered MLT/RGO/Polycaprolactone (PCL) composite scaffold was fabricated with beaded nanostructures to improve cell attachment and proliferation. It also exhibited stable mechanical properties by high elastic modulus and guaranteed structural integrity for nerve regeneration. The live/dead cell staining and cell counting kit assay were performed to evaluate the toxicity of the scaffold. JC-1 staining was carried out to assess the mitochondrial potential. The composite scaffold provided a biocompatible interface for cell viability and improved ATP production for energy supply. The scaffold improved the sensory and locomotor function recovery by walking track analysis and electrophysiological evaluation, reduced Schwann cell apoptosis and increased its proliferation. It further stimulated myelination and axonal outgrowth by enhancing S100β, myelin basic protein, β3-tubulin, and GAP43 levels. The findings demonstrated functional and morphological recovery by this biomimetic scaffold and indicated its potential for translational application.
Collapse
Affiliation(s)
- Huiquan Jiang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xu Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao Li
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Jin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiwen Yan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuanming Ouyang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
28
|
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: 34] [Impact Index Per Article: 17.0] [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
|
29
|
Rubey KM, Mukhitov AR, Nong J, Wu J, Krymskaya VP, Myerson JW, Worthen GS, Brenner JS. Nanoparticle-Induced Augmentation of Neutrophils' Phagocytosis of Bacteria. Front Pharmacol 2022; 13:923814. [PMID: 35860017 PMCID: PMC9289463 DOI: 10.3389/fphar.2022.923814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022] Open
Abstract
Despite the power of antibiotics, bacterial infections remain a major killer, due to antibiotic resistance and hosts with dysregulated immune systems. We and others have been developing drug-loaded nanoparticles that home to the sites of infection and inflammation via engineered tropism for neutrophils, the first-responder leukocytes in bacterial infections. Here, we examined how a member of a broad class of neutrophil-tropic nanoparticles affects neutrophil behavior, specifically questioning whether the nanoparticles attenuate an important function, bacterial phagocytosis. We found these nanoparticles actually augment phagocytosis of non-opsonized bacteria, increasing it by ∼50%. We showed this augmentation of phagocytosis is likely co-opting an evolved response, as opsonized bacteria also augment phagocytosis of non-opsonized bacteria. Enhancing phagocytosis of non-opsonized bacteria may prove particularly beneficial in two clinical situations: in hypocomplementemic patients (meaning low levels of the main bacterial opsonins, complement proteins, seen in conditions such as neonatal sepsis and liver failure) or for bacteria that are largely resistant to complement opsonization (e.g., Neisseria). Additionally, we observe that; 1) prior treatment with bacteria augments neutrophil uptake of neutrophil-tropic nanoparticles; 2) neutrophil-tropic nanoparticles colocalize with bacteria inside of neutrophils. The observation that neutrophil-tropic nanoparticles enhance neutrophil phagocytosis and localize with bacteria inside neutrophils suggests that these nanoparticles will serve as useful carriers for drugs to ameliorate bacterial diseases.
Collapse
Affiliation(s)
- Kathryn M Rubey
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alexander R Mukhitov
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jia Nong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Jichuan Wu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Vera P Krymskaya
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jacob W Myerson
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - G Scott Worthen
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jacob S Brenner
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
30
|
Srinivas M, Sharma P, Jhunjhunwala S. Phagocytic Uptake of Polymeric Particles by Immune Cells under Flow Conditions. Mol Pharm 2021; 18:4501-4510. [PMID: 34748349 DOI: 10.1021/acs.molpharmaceut.1c00698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Particles injected intravenously are thought to be cleared by macrophages residing in the liver and spleen, but they also encounter circulating immune cells. It remains to be established if the circulating cells can take up particles while flowing and if the uptake capacity is similar under static and flow conditions. Here, we use an in vitro peristaltic pump setup that mimics pulsatile blood flow to determine if immune cells take up particles under constant fluidic flow. We use polystyrene particles of varying sizes as the model of a polymeric particle for these studies. Our results show that the immune cells do phagocytose under flow conditions. We demonstrate that cell lines representing myeloid cells, primary human neutrophils, and monocytes take up submicrometer-sized particles at similar or better rates under flow compared to static conditions. Experiments with whole human blood show that, even under the crowding effects of red blood cells, neutrophils and monocytes take up particles while flowing. Together, these data suggest that circulating immune cells are likely to phagocytose intravenously injected particulates, which has implications for the design of particles to evade or target these cells.
Collapse
Affiliation(s)
- Megha Srinivas
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India.,Undergraduate Program, Indian Institute of Science, Bengaluru 560012, India
| | - Preeti Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Siddharth Jhunjhunwala
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
31
|
Li YJ, Wu JY, Liu J, Qiu X, Xu W, Tang T, Xiang DX. From blood to brain: blood cell-based biomimetic drug delivery systems. Drug Deliv 2021; 28:1214-1225. [PMID: 34142628 PMCID: PMC8259840 DOI: 10.1080/10717544.2021.1937384] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Brain drug delivery remains a major difficulty for several challenges including the blood-brain barrier, lesion spot targeting, and stability during circulation. Blood cells including erythrocytes, platelets, and various subpopulations of leukocytes have distinct features such as long-circulation, natural targeting, and chemotaxis. The development of biomimetic drug delivery systems based on blood cells for brain drug delivery is growing fast by using living cells, membrane coating nanotechnology, or cell membrane-derived nanovesicles. Blood cell-based vehicles are superior delivery systems for their engineering feasibility and versatile delivery ability of chemicals, proteins, and all kinds of nanoparticles. Here, we focus on advances of blood cell-based biomimetic carriers for from blood to brain drug delivery and discuss their translational challenges in the future.
Collapse
Affiliation(s)
- Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jihua Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiaohan Qiu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
32
|
Calvo A, Moreno E, Clemente U, Pérez E, Larrea E, Sanmartín C, Irache JM, Espuelas S. Changes in the nanoparticle uptake and distribution caused by an intramacrophagic parasitic infection. NANOSCALE 2021; 13:17486-17503. [PMID: 34651151 DOI: 10.1039/d1nr03797h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This study investigates if visceral leishmaniasis (VL) infection has some effects on the organ and cellular uptake and distribution of 100-200 nm near-infrared fluorescently labelled non-biodegradable polystyrene latex beads (PS NPs) or biodegradable polylactic-co-glycolic nanoparticles (PLGA NPs), as this parasitic infection produces morphological alterations in liver, spleen and bone marrow, organs highly involved in NP sequestration. The results showed that the magnitude of the effect was specific for each organ and type of NP. With the exception of the liver, the general trend was a decrease in NP organ and cellular uptake, mostly due to immune cell mobilization and/or weight organ gain, as vascular permeability was increased. Moreover, NPs redistributed among different phagocytic cells to adapt infection associated changes and cellular alterations. In the liver, it is noteworthy that only isolated Kuffer cells (KCs) captured NPs, whereas they were not taken up by KC forming granulomas. In the spleen, NPs redistributed from macrophages and dendritic cells towards B cells and inflammatory monocytes although they maintained their preferential accumulation in the marginal zone and red pulp. Comparatively, the infection rarely affected the NP cellular distribution in the bone marrow. NP cellular target changes in VL infection could affect their therapeutic efficacy and should be considered for more efficient drug delivery.
Collapse
Affiliation(s)
- Alba Calvo
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
| | - Esther Moreno
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- Navarra Institute for Health Research, IdisNA, Spain
| | - Unai Clemente
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
| | - Enma Pérez
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
| | - Esther Larrea
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Navarra Institute for Health Research, IdisNA, Spain
| | - Carmen Sanmartín
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- Navarra Institute for Health Research, IdisNA, Spain
| | - Juan Manuel Irache
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- Navarra Institute for Health Research, IdisNA, Spain
| | - Socorro Espuelas
- ISTUN Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- Chemistry and Pharmaceutical Technology Department, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- Navarra Institute for Health Research, IdisNA, Spain
| |
Collapse
|
33
|
Qiao Q, Liu X, Yang T, Cui K, Kong L, Yang C, Zhang Z. Nanomedicine for acute respiratory distress syndrome: The latest application, targeting strategy, and rational design. Acta Pharm Sin B 2021; 11:3060-3091. [PMID: 33977080 PMCID: PMC8102084 DOI: 10.1016/j.apsb.2021.04.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by the severe inflammation and destruction of the lung air-blood barrier, leading to irreversible and substantial respiratory function damage. Patients with coronavirus disease 2019 (COVID-19) have been encountered with a high risk of ARDS, underscoring the urgency for exploiting effective therapy. However, proper medications for ARDS are still lacking due to poor pharmacokinetics, non-specific side effects, inability to surmount pulmonary barrier, and inadequate management of heterogeneity. The increased lung permeability in the pathological environment of ARDS may contribute to nanoparticle-mediated passive targeting delivery. Nanomedicine has demonstrated unique advantages in solving the dilemma of ARDS drug therapy, which can address the shortcomings and limitations of traditional anti-inflammatory or antioxidant drug treatment. Through passive, active, or physicochemical targeting, nanocarriers can interact with lung epithelium/endothelium and inflammatory cells to reverse abnormal changes and restore homeostasis of the pulmonary environment, thereby showing good therapeutic activity and reduced toxicity. This article reviews the latest applications of nanomedicine in pre-clinical ARDS therapy, highlights the strategies for targeted treatment of lung inflammation, presents the innovative drug delivery systems, and provides inspiration for strengthening the therapeutic effect of nanomedicine-based treatment.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- AEC II, alveolar type II epithelial cells
- AM, alveolar macrophages
- ARDS, acute respiratory distress syndrome
- Acute lung injury
- Acute respiratory distress syndrome
- Anti-inflammatory therapy
- BALF, bronchoalveolar lavage fluid
- BSA, bovine serum albumin
- CD, cyclodextrin
- CLP, cecal ligation and perforation
- COVID-19
- COVID-19, coronavirus disease 2019
- DOPE, phosphatidylethanolamine
- DOTAP, 1-diolefin-3-trimethylaminopropane
- DOX, doxorubicin
- DPPC, dipalmitoylphosphatidylcholine
- Drug delivery
- ECM, extracellular matrix
- ELVIS, extravasation through leaky vasculature and subsequent inflammatory cell-mediated sequestration
- EPCs, endothelial progenitor cells
- EPR, enhanced permeability and retention
- EVs, extracellular vesicles
- EphA2, ephrin type-A receptor 2
- Esbp, E-selectin-binding peptide
- FcgR, Fcγ receptor
- GNP, peptide-gold nanoparticle
- H2O2, hydrogen peroxide
- HO-1, heme oxygenase-1
- ICAM-1, intercellular adhesion molecule-1
- IKK, IκB kinase
- IL, interleukin
- LPS, lipopolysaccharide
- MERS, Middle East respiratory syndrome
- MPMVECs, mouse pulmonary microvascular endothelial cells
- MPO, myeloperoxidase
- MSC, mesenchymal stem cells
- NAC, N-acetylcysteine
- NE, neutrophil elastase
- NETs, neutrophil extracellular traps
- NF-κB, nuclear factor-κB
- Nanomedicine
- PC, phosphatidylcholine
- PCB, poly(carboxybetaine)
- PDA, polydopamine
- PDE4, phosphodiesterase 4
- PECAM-1, platelet-endothelial cell adhesion molecule
- PEG, poly(ethylene glycol)
- PEI, polyetherimide
- PEVs, platelet-derived extracellular vesicles
- PLGA, poly(lactic-co-glycolic acid)
- PS-PEG, poly(styrene-b-ethylene glycol)
- Pathophysiologic feature
- RBC, red blood cells
- RBD, receptor-binding domains
- ROS, reactive oxygen species
- S1PLyase, sphingosine-1-phosphate lyase
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC1, syndecan-1
- SORT, selective organ targeting
- SP, surfactant protein
- Se, selenium
- Siglec, sialic acid-binding immunoglobulin-like lectin
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor-α
- TPP, triphenylphosphonium cation
- Targeting strategy
- YSA, YSAYPDSVPMMS
- cRGD, cyclic arginine glycine-d-aspartic acid
- iNOS, inducible nitric oxide synthase
- rSPANb, anti-rat SP-A nanobody
- scFv, single chain variable fragments
Collapse
Affiliation(s)
- Qi Qiao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kexin Cui
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
34
|
Wang F, Hou W, Xiao C, Hao Y, Su N, Deng Y, Wang J, Yu L, Xie JM, Xiong JW, Luo Y. Endothelial cell membrane-based biosurface for targeted delivery to acute injury: analysis of leukocyte-mediated nanoparticle transportation. NANOSCALE 2021; 13:14636-14643. [PMID: 34558568 DOI: 10.1039/d1nr04181a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.
Collapse
Affiliation(s)
- Fang Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Wenda Hou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Chenglu Xiao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Ni Su
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Yu Deng
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jieting Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Luying Yu
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America 02155
| |
Collapse
|
35
|
|
36
|
Bao J, Zhang Q, Duan T, Hu R, Tang J. The Fate of Nanoparticles In Vivo and the Strategy of Designing Stealth Nanoparticle for Drug Delivery. Curr Drug Targets 2021; 22:922-946. [PMID: 33461465 DOI: 10.2174/1389450122666210118105122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Nano-drug delivery systems (Nano-DDS) offer powerful advantages in drug delivery and targeted therapy for diseases. Compared to the traditional drug formulations, Nano-DDS can increase solubility, biocompatibility, and reduce off-targeted side effects of free drugs. However, they still have some disadvantages that pose a limitation in reaching their full potential in clinical use. Protein adsorption in blood, activation of the complement system, and subsequent sequestration by the mononuclear phagocyte system (MPS) consequently result in nanoparticles (NPs) to be rapidly cleared from circulation. Therefore, NPs have low drug delivery efficiency. So, it is important to develop stealth NPs for reducing bio-nano interaction. In this review, we first conclude the interaction between NPs and biological environments, such as blood proteins and MPS, and factors influencing each other. Next, we will summarize the new strategies to reduce NPs protein adsorption and uptake by the MPS based on current knowledge of the bio-nano interaction. Further directions will also be highlighted for the development of biomimetic stealth nano-delivery systems by combining targeted strategies for a better therapeutic effect.
Collapse
Affiliation(s)
- Jianwei Bao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qianqian Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Tijie Duan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rongfeng Hu
- key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Anhui "115" Xin'an Medicine Research & Development Innovation Team, Anhui Academy of Chinese Medicine, Hefei 230038, China
| | - Jihui Tang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
37
|
Jarai BM, Stillman Z, Bomb K, Kloxin AM, Fromen CA. Biomaterials-Based Opportunities to Engineer the Pulmonary Host Immune Response in COVID-19. ACS Biomater Sci Eng 2021; 7:1742-1764. [PMID: 33356134 PMCID: PMC7784663 DOI: 10.1021/acsbiomaterials.0c01287] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
Abstract
The COVID-19 pandemic caused by the global spread of the SARS-CoV-2 virus has led to a staggering number of deaths worldwide and significantly increased burden on healthcare as nations scramble to find mitigation strategies. While significant progress has been made in COVID-19 diagnostics and therapeutics, effective prevention and treatment options remain scarce. Because of the potential for the SARS-CoV-2 infections to cause systemic inflammation and multiple organ failure, it is imperative for the scientific community to evaluate therapeutic options aimed at modulating the causative host immune responses to prevent subsequent systemic complications. Harnessing decades of expertise in the use of natural and synthetic materials for biomedical applications, the biomaterials community has the potential to play an especially instrumental role in developing new strategies or repurposing existing tools to prevent or treat complications resulting from the COVID-19 pathology. Leveraging microparticle- and nanoparticle-based technology, especially in pulmonary delivery, biomaterials have demonstrated the ability to effectively modulate inflammation and may be well-suited for resolving SARS-CoV-2-induced effects. Here, we provide an overview of the SARS-CoV-2 virus infection and highlight current understanding of the host's pulmonary immune response and its contributions to disease severity and systemic inflammation. Comparing to frontline COVID-19 therapeutic options, we highlight the most significant untapped opportunities in immune engineering of the host response using biomaterials and particle technology, which have the potential to improve outcomes for COVID-19 patients, and identify areas needed for future investigations. We hope that this work will prompt preclinical and clinical investigations of promising biomaterials-based treatments to introduce new options for COVID-19 patients.
Collapse
Affiliation(s)
- Bader M. Jarai
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - Zachary Stillman
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - Kartik Bomb
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
38
|
Ahamad N, Kar A, Mehta S, Dewani M, Ravichandran V, Bhardwaj P, Sharma S, Banerjee R. Immunomodulatory nanosystems for treating inflammatory diseases. Biomaterials 2021; 274:120875. [PMID: 34010755 DOI: 10.1016/j.biomaterials.2021.120875] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/26/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023]
Abstract
Inflammatory disease (ID) is an umbrella term encompassing all illnesses involving chronic inflammation as the central manifestation of pathogenesis. These include, inflammatory bowel diseases, hepatitis, pulmonary disorders, atherosclerosis, myocardial infarction, pancreatitis, arthritis, periodontitis, psoriasis. The IDs create a severe burden on healthcare and significantly impact the global socio-economic balance. Unfortunately, the standard therapies that rely on a combination of anti-inflammatory and immunosuppressive agents are palliative and provide only short-term relief. In contrast, the emerging concept of immunomodulatory nanosystems (IMNs) has the potential to address the underlying causes and prevent reoccurrence, thereby, creating new opportunities for treating IDs. The IMNs offer exquisite ability to precisely modulate the immune system for a therapeutic advantage. The nano-sized dimension of IMNs allows them to efficiently infiltrate lymphatic drainage, interact with immune cells, and subsequently to undergo rapid endocytosis by hyperactive immune cells (HICs) at inflamed sites. Thus, IMNs serve to restore dysfunctional or HICs and alleviate the inflammation. We identified that different IMNs exert their immunomodulatory action via either of the seven mechanisms to modulate; cytokine production, cytokine neutralization, cellular infiltration, macrophage polarization, HICs growth inhibition, stimulating T-reg mediated tolerance and modulating oxidative-stress. In this article, we discussed representative examples of IMNs by highlighting their rationalization, design principle, and mechanism of action in context of treating various IDs. Lastly, we highlighted technical challenges in the application of IMNs and explored the future direction of research, which could potentially help to overcome those challenges.
Collapse
Affiliation(s)
- Nadim Ahamad
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Abhinanda Kar
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Sourabh Mehta
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India; IITB-Monash Research Academy IIT Bombay, Powai, Mumbai, 400076, India
| | - Mahima Dewani
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vasanthan Ravichandran
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Prateek Bhardwaj
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shivam Sharma
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rinti Banerjee
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
39
|
Zarubova J, Zhang X, Hoffman T, Hasani-Sadrabadi MM, Li S. Biomaterial-based immunoengineering to fight COVID-19 and infectious diseases. MATTER 2021; 4:1528-1554. [PMID: 33723531 PMCID: PMC7942141 DOI: 10.1016/j.matt.2021.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Infection by SARS-CoV-2 virus often induces the dysregulation of immune responses, tissue damage, and blood clotting. Engineered biomaterials from the nano- to the macroscale can provide targeted drug delivery, controlled drug release, local immunomodulation, enhanced immunity, and other desirable functions to coordinate appropriate immune responses and to repair tissues. Based on the understanding of COVID-19 disease progression and immune responses to SARS-CoV-2, we discuss possible immunotherapeutic strategies and highlight biomaterial approaches from the perspectives of preventive immunization, therapeutic immunomodulation, and tissue healing and regeneration. Successful development of biomaterial platforms for immunization and immunomodulation will not only benefit COVID-19 patients, but also have broad applications for a variety of infectious diseases.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Xuexiang Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Ong YR, De Rose R, Johnston APR. In Vivo Quantification of Nanoparticle Association with Immune Cell Subsets in Blood. Adv Healthc Mater 2021; 10:e2002160. [PMID: 33644997 DOI: 10.1002/adhm.202002160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/27/2021] [Indexed: 12/17/2022]
Abstract
Nanoparticles offer great promise for more effective drug delivery. However, their particulate nature typically results in rapid systemic clearance by immune cells in blood. Currently, to understand these interactions, nanoparticle association is probed ex vivo with whole blood. While ex vivo assays give important information about the relative cell association, they do not consider changes in immune cell homeostasis or the complex mixing behavior that occurs in vivo. To address this, a nanoparticle in vivo immune-cell association assay is developed to study the in vivo association of unmodified and poly(ethylene glycol) modified liposomes with immune cells, and compared this to the ex vivo association in static whole blood. In vivo, it is observed that neutrophils play a significantly greater role in nanoparticle binding than suggested by ex vivo assays. The increased influence of neutrophils in vivo is largely due to a significant increase in number of circulating neutrophils after intravenous injection. Conversely, the number of circulating monocytes significantly decreased after intravenous injection, leading to significantly less total association of liposomes to monocytes compared to ex vivo. This novel in vivo immune cell binding assay sheds new light on the fate of nanoparticles following intravenous delivery.
Collapse
Affiliation(s)
- Yih Rue Ong
- Monash Institute of Pharmaceutical Sciences Monash University 399 Royal Parade Parkville Victoria 3052 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash University Parkville Victoria 3052 Australia
| | - Robert De Rose
- Monash Institute of Pharmaceutical Sciences Monash University 399 Royal Parade Parkville Victoria 3052 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash University Parkville Victoria 3052 Australia
| | - Angus P. R. Johnston
- Monash Institute of Pharmaceutical Sciences Monash University 399 Royal Parade Parkville Victoria 3052 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash University Parkville Victoria 3052 Australia
| |
Collapse
|
41
|
Vishnevskiy DA, Garanina AS, Chernysheva AA, Chekhonin VP, Naumenko VA. Neutrophil and Nanoparticles Delivery to Tumor: Is It Going to Carry That Weight? Adv Healthc Mater 2021; 10:e2002071. [PMID: 33734620 DOI: 10.1002/adhm.202002071] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/02/2021] [Indexed: 12/15/2022]
Abstract
The application of cell carriers for transporting nanodrugs to the tumor draws much attention as the alternative to the passive drug delivery. In this concept, the neutrophil (NΦ) is of special interest as this cell is able to uptake nanoparticles (NPs) and cross the vascular barrier in response to tumor signaling. There is a growing body of literature describing NP-NΦ interactions in vitro and in vivo that demonstrates the opportunity of using these cells to improve the efficacy of cancer therapy. However, a number of conceptual and technical issues need to be resolved for translating the technology into clinics. The current review summarizes the recent advances and challenges associated with NP-NΦ interactions, with the special focus on the complex interplay between the NP internalization pathways and the modulation of NΦ activity, and its potential consequences for nanodrug delivery.
Collapse
Affiliation(s)
- Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
- N. I Pirogov Russian National Research Medical University Ulitsa Ostrovityanova, 1 Moscow 117997 Russia
| | - Anastasiia S. Garanina
- National University of Science and Technology (MISIS) Leninskiy Prospekt, 4 Moscow 119049 Russia
| | - Anastasia A. Chernysheva
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
- N. I Pirogov Russian National Research Medical University Ulitsa Ostrovityanova, 1 Moscow 117997 Russia
| | - Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
| |
Collapse
|
42
|
Uchiyama MK, Hebeda CB, Sandri S, Paula-Silva MD, Romano M, Cardoso RM, Toma SH, Araki K, Farsky SH. In vivo evaluation of toxicity and anti-inflammatory activity of iron oxide nanoparticles conjugated with ibuprofen. Nanomedicine (Lond) 2021; 16:741-758. [PMID: 33856243 DOI: 10.2217/nnm-2020-0459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The low solubility and consequent poor bioavailability of ibuprofen (IBU) is a major drawback that can be overcome by anchoring IBU on ultrasmall superparamagnetic iron oxide nanoparticles (USPIONs) as effective multifunctional carriers for drug delivery. Methods: USPIONs were conjugated with glycerol phosphate (USPION-GP) and also co-conjugated with IBU (USPION-GP/IBU), and their in vivo toxicity and anti-inflammatory effects investigated. Phosphate buffer saline (control), IBU, USPION-GP and USPION-GP/IBU were intravenously administered 15 min before lipopolysaccharide-induced peritonitis in male Balb/c mice. Results: 4 h later, USPION bioconjugates did not appear to have caused toxicity to blood leukocytes or caused alterations in the spleen, liver or kidneys. Also, they inhibited lipopolysaccharide-induced neutrophil mobilization into the peritoneum. Conclusion: The absence of systemic toxicity and the unexpected anti-inflammatory action of USPION bioconjugates indicates that they could be a novel and effective approach to administer IBU and warrant further investigation.
Collapse
Affiliation(s)
- Mayara K Uchiyama
- Department of Fundamental Chemistry, Laboratory of Supramolecular Chemistry & Nanotechnology, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, São Paulo, 05508-000, Brazil
| | - Cristina B Hebeda
- Department of Clinical & Toxicological Analysis, Laboratory of Experimental Toxicology, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, São Paulo, 05508-000, Brazil
| | - Silvana Sandri
- Department of Clinical & Toxicological Analysis, Laboratory of Experimental Toxicology, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, São Paulo, 05508-000, Brazil
| | - Marina de Paula-Silva
- Department of Clinical & Toxicological Analysis, Laboratory of Experimental Toxicology, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, São Paulo, 05508-000, Brazil
| | - Mariana Romano
- Department of Fundamental Chemistry, Laboratory of Supramolecular Chemistry & Nanotechnology, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, São Paulo, 05508-000, Brazil
| | - Roberta M Cardoso
- Department of Fundamental Chemistry, Laboratory of Supramolecular Chemistry & Nanotechnology, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, São Paulo, 05508-000, Brazil
| | - Sergio H Toma
- Department of Fundamental Chemistry, Laboratory of Supramolecular Chemistry & Nanotechnology, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, São Paulo, 05508-000, Brazil
| | - Koiti Araki
- Department of Fundamental Chemistry, Laboratory of Supramolecular Chemistry & Nanotechnology, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, São Paulo, 05508-000, Brazil
| | - Sandra Hp Farsky
- Department of Clinical & Toxicological Analysis, Laboratory of Experimental Toxicology, Faculty of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, São Paulo, 05508-000, Brazil
| |
Collapse
|
43
|
Fish MB, Banka AL, Braunreuther M, Fromen CA, Kelley WJ, Lee J, Adili R, Holinstat M, Eniola-Adefeso O. Deformable microparticles for shuttling nanoparticles to the vascular wall. SCIENCE ADVANCES 2021; 7:eabe0143. [PMID: 33883129 PMCID: PMC8059934 DOI: 10.1126/sciadv.abe0143] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 03/08/2021] [Indexed: 05/11/2023]
Abstract
Vascular-targeted drug carriers must localize to the wall (i.e., marginate) and adhere to a diseased endothelium to achieve clinical utility. The particle size has been reported as a critical physical property prescribing particle margination in vitro and in vivo blood flows. Different transport process steps yield conflicting requirements-microparticles are optimal for margination, but nanoparticles are better for intracellular or tissue delivery. Here, we evaluate deformable hydrogel microparticles as carriers for transporting nanoparticles to a diseased vascular wall. Depending on microparticle modulus, nanoparticle-loaded poly(ethylene glycol)-based hydrogel microparticles delivered significantly more 50-nm nanoparticles to the vessel wall than freely injected nanoparticles alone, resulting in >3000% delivery increase. This work demonstrates the benefit of optimizing microparticles' efficient margination to enhance nanocarriers' transport to the vascular wall.
Collapse
Affiliation(s)
- Margaret B Fish
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alison L Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret Braunreuther
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Catherine A Fromen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - William J Kelley
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Lee
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
44
|
Rong J, He Y, Tang J, Qiao R, Lin S. "Fishing" nano-bio interactions at the key biological barriers. NANOSCALE 2021; 13:5954-5964. [PMID: 33734277 DOI: 10.1039/d1nr00328c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Understanding nano-bio interactions is pivotal to the safe implementation of nanotechnology for both biological and environmental applications. Zebrafish as a model organism provides unique opportunities to dissect nano-bio interactions occurring at different biological barriers. In this review, we focus on four key biological barriers, namely cell membrane, blood-brain barrier (BBB), skin and gill epithelia, and gastrointestinal tract (GIT), and highlight recent advancement achieved by using zebrafish to conduct both visualized observations and mechanistic investigations on a diversity of nano-bio interactions.
Collapse
Affiliation(s)
- Jinyu Rong
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Naumenko VA, Vodopyanov SS, Vlasova KY, Potashnikova DM, Melnikov PA, Vishnevskiy DA, Garanina AS, Valikhov MP, Lipatova AV, Chekhonin VP, Majouga AG, Abakumov MA. Intravital imaging of liposome behavior upon repeated administration: A step towards the development of liposomal companion diagnostic for cancer nanotherapy. J Control Release 2021; 330:244-256. [DOI: 10.1016/j.jconrel.2020.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/29/2020] [Accepted: 12/11/2020] [Indexed: 01/04/2023]
|
47
|
Zhang Y, Hughes KR, Raghani RM, Ma J, Orbach S, Jeruss JS, Shea LD. Cargo-free immunomodulatory nanoparticles combined with anti-PD-1 antibody for treating metastatic breast cancer. Biomaterials 2021; 269:120666. [PMID: 33461057 DOI: 10.1016/j.biomaterials.2021.120666] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
The presence of immunosuppressive innate immune cells such as myeloid derived suppressor cells (MDSCs), Ly6C-high monocytes, and tumor-associated macrophages (TAMs) at a tumor can inhibit effector T cell and NK cell function. Immune checkpoint blockade using anti-PD-1 antibody aims to overcome the immune suppressive environment, yet only a fraction of patients responds. Herein, we test the hypothesis that cargo-free PLG nanoparticles administered intravenously can divert circulating immune cells from the tumor microenvironment to enhance the efficacy of anti-PD-1 immunotherapy in the 4T1 mouse model of metastatic triple-negative breast cancer. In vitro studies demonstrate that these nanoparticles decrease the expression of MCP-1 by 5-fold and increase the expression of TNF-α by more than 2-fold upon uptake by innate immune cells. Intravenous administration of particles results in internalization by MDSCs and monocytes, with particles detected in the liver, lung, spleen, and primary tumor. Nanoparticle delivery decreased the abundance of MDSCs in circulation and in the lung, the latter being the primary metastatic site. Combined with anti-PD-1 antibody, nanoparticles significantly slowed tumor growth and resulted in a survival benefit. Gene expression analysis by GSEA indicated inflammatory myeloid cell pathways were downregulated in the lung and upregulated in the spleen and tumor. Upregulation of extrinsic apoptotic pathways was also observed in the primary tumor. Collectively, these results demonstrate that cargo-free PLG nanoparticles can reprogram immune cell responses and alter the tumor microenvironment in vivo to overcome the local immune suppression attributed to myeloid cells and enhance the efficacy of anti-PD-1 therapy.
Collapse
Affiliation(s)
- Yining Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ravi M Raghani
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeffrey Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sophia Orbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lonnie D Shea
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
48
|
Neutrophils and Macrophages as Targets for Development of Nanotherapeutics in Inflammatory Diseases. Pharmaceutics 2020; 12:pharmaceutics12121222. [PMID: 33348630 PMCID: PMC7766591 DOI: 10.3390/pharmaceutics12121222] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Neutrophils and macrophages are major components of innate systems, playing central roles in inflammation responses to infections and tissue injury. If they are out of control, inflammation responses can cause the pathogenesis of a wide range of diseases, such as inflammatory disorders and autoimmune diseases. Precisely regulating the functions of neutrophils and macrophages in vivo is a potential strategy to develop immunotherapies to treat inflammatory diseases. Advances in nanotechnology have enabled us to design nanoparticles capable of targeting neutrophils or macrophages in vivo. This review discusses the current status of how nanoparticles specifically target neutrophils or macrophages and how they manipulate leukocyte functions to inhibit their activation for inflammation resolution or to restore their defense ability for pathogen clearance. Finally, we present a novel concept of hijacking leukocytes to deliver nanotherapeutics across the blood vessel barrier. This review highlights the challenges and opportunities in developing nanotherapeutics to target leukocytes for improved treatment of inflammatory diseases.
Collapse
|
49
|
Garcia G, Kim MH, Morikis VA, Simon SI. Neutrophil Inflammatory Response Is Downregulated by Uptake of Superparamagnetic Iron Oxide Nanoparticle Therapeutics. Front Immunol 2020; 11:571489. [PMID: 33362760 PMCID: PMC7757401 DOI: 10.3389/fimmu.2020.571489] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are employed as diagnostics and therapeutics following intravenous delivery for the treatment of iron deficiency anemia (IDA) in adult patients with chronic kidney failure. Neutrophils are the first defense against blood borne foreign insult and recruit to vascular sites of inflammation via a sequential process that is characterized by adhesive capture, rolling, and shear resistant arrest. A primary chemotactic agonist presented on the glycocalyx of inflamed endothelium is IL-8, which upon binding to its cognate membrane receptor (CXCR1/2) activates a suite of responses in neutrophils. An early response is degranulation with accompanying upregulation of β2-integrin (CD11/CD18) and shedding of L-selectin (CD62L) receptors, which exert differential effects on the efficiency of endothelial recruitment. Feraheme is an FDA approved SPION treatment for IDA, but its effect on the innate immune response of neutrophils during inflammation has not been reported. Here, we studied the immunomodulatory effects of Feraheme on neutrophils freshly isolated from healthy human subjects and stimulated in suspension or on inflammatory mimetic substrates with IL-8. Cells treated with Feraheme exhibited reduced sensitivity to stimulation with IL-8, indicated by reduced upregulation of membrane CD11b/CD18 receptors, high affinity (HA) CD18, and shedding of CD62L. Feraheme also inhibited N-formyl-Met-Leu-Phe (fMLP) induced reactive oxygen species production. Neutrophil rolling, arrest, and migration was assessed in vascular mimetic microfluidic channels coated with E-selectin and ICAM-1 to simulate inflamed endothelium. Neutrophils exposed to Feraheme rolled faster on E-selectin and arrested less frequently on ICAM-1, in a manner dependent upon SPION concentration. Subsequent neutrophil shape change, and migration were also significantly inhibited in the presence of Feraheme. Lastly, Feraheme accelerated clearance of cytosolic calcium flux following IL-8 stimulation. We conclude that uptake of Feraheme by neutrophils inhibits chemotactic activation and downregulates normal rolling to arrest under shear flow. The mechanism involves increased calcium clearance following chemotactic activation, which may diminish the efficiency of recruitment from the circulation at vascular sites of inflammation.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Min-Ho Kim
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Vasilios Aris Morikis
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Scott I. Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
50
|
Shirazi J, Donzanti MJ, Nelson KM, Zurakowski R, Fromen CA, Gleghorn JP. Significant Unresolved Questions and Opportunities for Bioengineering in Understanding and Treating COVID-19 Disease Progression. Cell Mol Bioeng 2020; 13:259-284. [PMID: 32837585 PMCID: PMC7384395 DOI: 10.1007/s12195-020-00637-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 is a disease that manifests itself in a multitude of ways across a wide range of tissues. Many factors are involved, and though impressive strides have been made in studying this novel disease in a very short time, there is still a great deal that is unknown about how the virus functions. Clinical data has been crucial for providing information on COVID-19 progression and determining risk factors. However, the mechanisms leading to the multi-tissue pathology are yet to be fully established. Although insights from SARS-CoV-1 and MERS-CoV have been valuable, it is clear that SARS-CoV-2 is different and merits its own extensive studies. In this review, we highlight unresolved questions surrounding this virus including the temporal immune dynamics, infection of non-pulmonary tissue, early life exposure, and the role of circadian rhythms. Risk factors such as sex and exposure to pollutants are also explored followed by a discussion of ways in which bioengineering approaches can be employed to help understand COVID-19. The use of sophisticated in vitro models can be employed to interrogate intercellular interactions and also to tease apart effects of the virus itself from the resulting immune response. Additionally, spatiotemporal information can be gleaned from these models to learn more about the dynamics of the virus and COVID-19 progression. Application of advanced tissue and organ system models into COVID-19 research can result in more nuanced insight into the mechanisms underlying this condition and elucidate strategies to combat its effects.
Collapse
Affiliation(s)
- Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Michael J. Donzanti
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Ryan Zurakowski
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| |
Collapse
|