Self-therapeutic metal-based nanoparticles for treating inflammatory diseases

Acta Pharmaceutica Sinica B - Tập 13 - Trang 1847-1865 - 2023
Ruifang Han1, Yu Xiao1, Qianqian Bai1, Chung Hang Jonathan Choi1
1Department of Biomedical Engineering, the Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China

Tài liệu tham khảo

Chen, 2017, Inflammatory responses and inflammation-associated diseases in organs, Oncotarget, 9, 7204, 10.18632/oncotarget.23208 Hirayama, 2017, The phagocytic function of macrophage-enforcing innate immunity and tissue homeostasis, Int J Mol Sci, 19, 92, 10.3390/ijms19010092 Li, 2022, Bioactive materials promote wound healing through modulation of cell behaviors, Adv Sci (Weinh), 9 Ryan, 1977, Acute inflammation. A review, Am J Pathol, 86, 183 Furman, 2019, Chronic inflammation in the etiology of disease across the life span, Nat Med, 25, 1822, 10.1038/s41591-019-0675-0 Hawkins, 2019, Detection, identification, and quantification of oxidative protein modifications, J Biol Chem, 294, 19683, 10.1074/jbc.REV119.006217 Zielinski, 2017, Lipid peroxidation: kinetics, mechanisms, and products, J Org Chem, 82, 2817, 10.1021/acs.joc.7b00152 Chetyrkin, 2011, Glucose autoxidation induces functional damage to proteins via modification of critical arginine residues, Biochemistry, 50, 6102, 10.1021/bi200757d Blaser, 2016, TNF and ROS crosstalk in inflammation, Trends Cell Biol, 26, 249, 10.1016/j.tcb.2015.12.002 Giacco, 2010, Oxidative stress and diabetic complications, Circ Res, 107, 1058, 10.1161/CIRCRESAHA.110.223545 Sahiner, 2011, Oxidative stress in asthma, World Allergy Organ J, 4, 151, 10.1097/WOX.0b013e318232389e Quiñonez-Flores, 2016, Oxidative stress relevance in the pathogenesis of the rheumatoid arthritis: a systematic review, BioMed Res Int, 2016, 10.1155/2016/6097417 Morgan, 2011, Crosstalk of reactive oxygen species and NF-κB signaling, Cell Res, 21, 103, 10.1038/cr.2010.178 Son, 2011, Mitogen-activated protein kinases and reactive oxygen species: how can ROS activate MAPK pathways?, J Signal Transduct, 2011, 10.1155/2011/792639 Banerjee, 2017, JAK–STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects, Drugs, 77, 521, 10.1007/s40265-017-0701-9 Liongue, 2013, Evolution of the JAK-STAT pathway, JAK-STAT, 2, 10.4161/jkst.22756 Clark, 2014, Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases, J Med Chem, 57, 5023, 10.1021/jm401490p Billing, 2019, Robustness and information transfer within IL-6-induced JAK/STAT signalling, Commun Biol, 2, 27, 10.1038/s42003-018-0259-4 Girard, 2009, Role of perinatal inflammation in cerebral palsy, Pediatr Neurol, 40, 168, 10.1016/j.pediatrneurol.2008.09.016 Sun, 2013, Regulation of nuclear factor-κB in autoimmunity, Trends Immunol, 34, 282, 10.1016/j.it.2013.01.004 Pasparakis, 2006, Dissection of the NF-kappaB signalling cascade in transgenic and knockout mice, Cell Death Differ, 13, 861, 10.1038/sj.cdd.4401870 Basak, 2007, A fourth IkappaB protein within the NF-kappaB signaling module, Cell, 128, 369, 10.1016/j.cell.2006.12.033 Ma, 2016, Crosstalk between Wnt/β-Catenin and NF-κB signaling pathway during inflammation, Front Immunol, 7, 378, 10.3389/fimmu.2016.00378 Atreya, 2008, NF-κB in inflammatory bowel disease, J Intern Med, 263, 591, 10.1111/j.1365-2796.2008.01953.x Edwards, 2009, Targeting the NF-κB pathway in asthma and chronic obstructive pulmonary disease, Pharmacol Ther, 121, 1, 10.1016/j.pharmthera.2008.09.003 Roman-Blas, 2006, NF-κB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis, Osteoarthritis Cartilage, 14, 839, 10.1016/j.joca.2006.04.008 Mc Guire, 2013, Nuclear factor kappa B (NF-κB) in multiple sclerosis pathology, Trends Mol Med, 19, 604, 10.1016/j.molmed.2013.08.001 Gareus, 2008, Endothelial cell-specific NF-κB inhibition protects mice from atherosclerosis, Cell Metabol, 8, 372, 10.1016/j.cmet.2008.08.016 Orr, 2005, The subendothelial extracellular matrix modulates NF-κB activation by flow: a potential role in atherosclerosis, J Cell Biol, 169, 191, 10.1083/jcb.200410073 Thalhamer, 2008, MAPKs and their relevance to arthritis and inflammation, Rheumatology (Oxford), 47, 409, 10.1093/rheumatology/kem297 Herlaar, 1999, p38 MAPK signalling cascades in inflammatory disease, Mol Med Today, 5, 439, 10.1016/S1357-4310(99)01544-0 Barnes, 2006, Novel signal transduction modulators for the treatment of airway diseases, Pharmacol Ther, 109, 238, 10.1016/j.pharmthera.2005.08.001 Haddad, 2001, Role of p38 MAP kinase in LPS-induced airway inflammation in the rat, Br J Pharmacol, 132, 1715, 10.1038/sj.bjp.0704022 Scaldaferri, 2009, The role of MAPK in governing lymphocyte adhesion to and migration across the microvasculature in inflammatory bowel disease, Eur J Immunol, 39, 290, 10.1002/eji.200838316 Sun, 2016, The mitogen-activated protein kinase (MAPK) signaling pathway as a discovery target in stroke, J Mol Neurosci, 59, 90, 10.1007/s12031-016-0717-8 Buckley, 2013, The resolution of inflammation, Nat Rev Immunol, 13, 59, 10.1038/nri3362 Ho, 2006, The efficacy of corticosteroid therapy in inflammatory bowel disease: analysis of a 5-year UK inception cohort, Aliment Pharmacol Ther, 24, 319, 10.1111/j.1365-2036.2006.02974.x Dhasmana, 2014, The effectiveness of nonsteroidal anti-inflammatory agents in the treatment of pelvic inflammatory disease: a systematic review, Syst Rev, 3, 79, 10.1186/2046-4053-3-79 Jabs, 2000, Guidelines for the use of immunosuppressive drugs in patients with ocular inflammatory disorders: recommendations of an expert panel, Am J Ophthalmol, 130, 492, 10.1016/S0002-9394(00)00659-0 Ivanenkov, 2008, New approaches to the treatment of inflammatory disease, Drugs R D, 9, 397, 10.2165/0126839-200809060-00005 Baugh, 2001, Mechanisms for modulating TNF alpha in immune and inflammatory disease, Curr Opin Drug Discov Dev, 4, 635 Kamata, 2020, Efficacy and safety of biologics for psoriasis and psoriatic arthritis and their impact on comorbidities: a literature review, Int J Mol Sci, 21, 1690, 10.3390/ijms21051690 Li, 2020, Comparative risk of serious infections among real-world users of biologics for psoriasis or psoriatic arthritis, Ann Rheum Dis, 79, 285, 10.1136/annrheumdis-2019-216102 Su, 2022, Polymer nanotherapeutics to correct autoimmunity, J Control Release, 343, 152, 10.1016/j.jconrel.2021.12.036 Choi, 2007, Renal clearance of quantum dots, Nat Biotechnol, 25, 1165, 10.1038/nbt1340 Beltrán-Gracia, 2019, Nanomedicine review: clinical developments in liposomal applications, Cancer Nano, 10, 11, 10.1186/s12645-019-0055-y van der Valk, 2015, Prednisolone-containing liposomes accumulate in human atherosclerotic macrophages upon intravenous administration, Nanomedicine, 11, 1039, 10.1016/j.nano.2015.02.021 Kulkarni, 2017, Self-assembled lipid cubic phase and cubosomes for the delivery of aspirin as a model drug, Langmuir, 33, 9907, 10.1021/acs.langmuir.7b02486 Guimarães, 2020, Increased encapsulation efficiency of methotrexate in liposomes for rheumatoid arthritis therapy, Biomedicine, 8, 630 Kim, 2018, Nanoparticle-assisted transcutaneous delivery of a signal transducer and activator of transcription 3-inhibiting peptide ameliorates psoriasis-like skin inflammation, ACS Nano, 12, 6904, 10.1021/acsnano.8b02330 Veiga, 2019, Leukocyte-specific siRNA delivery revealing IRF8 as a potential anti-inflammatory target, J Control Release, 313, 33, 10.1016/j.jconrel.2019.10.001 He, 2021, Intra-articular injection of lornoxicam and microRNA-140 co-loaded cationic liposomes enhanced the therapeutic treatment of experimental osteoarthritis, AAPS PharmSciTech, 23, 9, 10.1208/s12249-021-02149-w Blanco, 2015, Principles of nanoparticle design for overcoming biological barriers to drug delivery, Nat Biotechnol, 33, 941, 10.1038/nbt.3330 Han, 2021, Alkyl-terminated gold nanoparticles as a self-therapeutic treatment for psoriasis, Nano Lett, 21, 8723, 10.1021/acs.nanolett.1c02899 Ohta, 2020, Investigating the optimum size of nanoparticles for their delivery into the brain assisted by focused ultrasound-induced blood–brain barrier opening, Sci Rep, 10, 10.1038/s41598-020-75253-9 Liu, 2021, Dopamine receptor-mediated binding and cellular uptake of polydopamine-coated nanoparticles, ACS Nano, 15, 13871, 10.1021/acsnano.1c06081 Sau, 2010, Properties and applications of colloidal nonspherical noble metal nanoparticles, Adv Mater, 22, 1805, 10.1002/adma.200902557 Yang, 2018, Promoting intracellular delivery of sub-25 nm nanoparticles via defined levels of compression, Nanoscale, 10, 15090, 10.1039/C8NR04927K Yang, 2016, Mechanism for the cellular uptake of targeted gold nanorods of defined aspect ratios, Small, 12, 5178, 10.1002/smll.201601483 Yin, 2021, Sub-10 nm substrate roughness promotes the cellular uptake of nanoparticles by upregulating endocytosis-related genes, Nano Lett, 21, 1839, 10.1021/acs.nanolett.0c04932 Ho, 2021, Effect of surface modification with hydrocarbyl groups on the exocytosis of nanoparticles, Biochemistry, 60, 1019, 10.1021/acs.biochem.0c00631 Choi, 2011, Targeting kidney mesangium by nanoparticles of defined size, Proc Natl Acad Sci U S A, 108, 6656, 10.1073/pnas.1103573108 Ho, 2022, Mammalian cells exocytose alkylated gold nanoparticles via extracellular vesicles, ACS Nano, 16, 2032, 10.1021/acsnano.1c07418 Agarwal, 2019, Anti-inflammatory mechanism of various metal and metal oxide nanoparticles synthesized using plant extracts: a review, Biomed Pharmacother, 109, 2561, 10.1016/j.biopha.2018.11.116 Adeel, 2020, Self-therapeutic nanomaterials for cancer therapy: a review, ACS Appl Nano Mater, 3, 4962, 10.1021/acsanm.0c00762 Shan, 2022, Current approaches of nanomedicines in the market and various stage of clinical translation, Acta Pharm Sin B, 12, 3028, 10.1016/j.apsb.2022.02.025 Ho, 2017, Effect of alkylation on the cellular uptake of polyethylene glycol-coated gold nanoparticles, ACS Nano, 11, 6085, 10.1021/acsnano.7b02044 Özcan, 2020, Nanoparticle-coupled topical methotrexate can normalize immune responses and induce tissue remodeling in psoriasis, J Invest Dermatol, 140, 1003, 10.1016/j.jid.2019.09.018 Ahn, 2013, Gold nanoparticle contrast agents in advanced X-ray imaging technologies, Molecules, 18, 5858, 10.3390/molecules18055858 Choi, 2019, Dopamine-mediated assembly of citrate-capped plasmonic nanoparticles into stable core-shell nanoworms for intracellular applications, ACS Nano, 13, 5864, 10.1021/acsnano.9b01591 Dai, 2021, Immobilising hairpin DNA-conjugated distyryl boron dipyrromethene on gold@polydopamine core–shell nanorods for microRNA detection and microRNA-mediated photodynamic therapy, Nanoscale, 13, 6499, 10.1039/D0NR09135A Dos Santos Haupenthal, 2020, Effects of treatment with gold nanoparticles in a model of acute pulmonary inflammation induced by lipopolysaccharide, J Biomed Mater Res A, 108, 103, 10.1002/jbm.a.36796 van der Fits, 2009, Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis, J Immunol, 182, 5836, 10.4049/jimmunol.0802999 Abdel-Hakem, 2022, Therapeutic outcomes and biodistribution of gold nanoparticles in collagen-induced arthritis animal model, J Drug Deliv Sci Technol, 67 Liu, 2020, Ultrasmall copper-based nanoparticles for reactive oxygen species scavenging and alleviation of inflammation related diseases, Nat Commun, 11, 2788, 10.1038/s41467-020-16544-7 Zhu, 2019, Platinum nanoparticles as a therapeutic agent against dextran sodium sulfate-induced colitis in mice, Int J Nanomed, 14, 8361, 10.2147/IJN.S210655 Zhang, 2020, Ultrasmall platinum nanozymes as broad-spectrum antioxidants for theranostic application in acute kidney injury, Chem Eng J, 409 Jeong, 2019, Ceria nanoparticles fabricated with 6-aminohexanoic acid that overcome systemic inflammatory response syndrome, Adv Healthc Mater, 8, 10.1002/adhm.201801548 Kim, 2021, Ultrasmall antioxidant cerium oxide nanoparticles for regulation of acute inflammation, ACS Appl Mater Interfaces, 13, 60852, 10.1021/acsami.1c16126 Li, 2020, Dual detoxification and inflammatory regulation by ceria nanozymes for drug-induced liver injury therapy, Nano Today, 35, 10.1016/j.nantod.2020.100925 Kalashnikova, 2020, Ceria-based nanotheranostic agent for rheumatoid arthritis, Theranostics, 10, 11863, 10.7150/thno.49069 Abdulmalek, 2021, Protective effect of natural antioxidant, curcumin nanoparticles, and zinc oxide nanoparticles against type 2 diabetes-promoted hippocampal neurotoxicity in rats, Pharmaceutics, 13, 1937, 10.3390/pharmaceutics13111937 Abdulmalek, 2021, Ameliorative effect of curcumin and zinc oxide nanoparticles on multiple mechanisms in obese rats with induced type 2 diabetes, Sci Rep, 11, 10.1038/s41598-021-00108-w Lin, 2022, Bioengineered zinc oxide nanoparticle-loaded hydrogel for combinative treatment of spinal cord transection, Front Bioeng Biotechnol, 9, 10.3389/fbioe.2021.796361 Chakrabartty, 2019, Inhibition of snake venom induced sterile inflammation and PLA2 activity by titanium dioxide nanoparticles in experimental animals, Sci Rep, 9, 10.1038/s41598-019-47557-y Miao, 2020, Ultrasmall rhodium nanozyme with RONS scavenging and photothermal activities for anti-inflammation and antitumor theranostics of colon diseases, Nano Lett, 20, 3079, 10.1021/acs.nanolett.9b05035 Wu, 2021, Multi-enzymatic activities of ultrasmall ruthenium oxide for anti-inflammation and neuroprotection, Chem Eng J, 411, 10.1016/j.cej.2021.128543 Li, 2021, Synergistic effects of silver nanoparticles and cisplatin in combating inflammation and hyperplasia of airway stents, Bioact Mater, 9, 266, 10.1016/j.bioactmat.2021.07.029 Kim, 2015, Zinc oxide nanoparticles suppress LPS-induced NF-κB activation by inducing A20, a negative regulator of NF-κB, in RAW 264.7 macrophages, J Nanosci Nanotechnol, 15, 6509, 10.1166/jnn.2015.10319 Selvaraj, 2015, Inhibition of MAP kinase/NF-κB mediated signaling and attenuation of lipopolysaccharide induced severe sepsis by cerium oxide nanoparticles, Biomaterials, 59, 160, 10.1016/j.biomaterials.2015.04.025 Sumbayev, 2013, Gold nanoparticles downregulate interleukin-1β-induced pro-inflammatory responses, Small, 9, 472, 10.1002/smll.201201528 Kingston, 2016, Selective inhibitory effects of 50-nm gold nanoparticles on mouse macrophage and spleen cells, J Immunot, 13, 198, 10.3109/1547691X.2015.1035819 de Carvalho, 2018, Spherical neutral gold nanoparticles improve anti-inflammatory response, oxidative stress and fibrosis in alcohol-methamphetamine-induced liver injury in rats, Int J Pharm, 548, 1, 10.1016/j.ijpharm.2018.06.008 Lopez-Chaves, 2018, Gold nanoparticles: distribution, bioaccumulation and toxicity. In vitro and in vivo studies, Nanomedicine, 14, 1, 10.1016/j.nano.2017.08.011 Nedoszutko, 2014, Chemokines and cytokines network in the pathogenesis of the inflammatory skin diseases: atopic dermatitis, psoriasis and skin mastocytosis, Postepy Dermatol Alergol, 31, 84, 10.5114/pdia.2014.40920 Lee, 2012, Pathophysiology of chemokines and chemokine receptors in dermatological science: a focus on psoriasis and cutaneous T-cell lymphoma, Dermatol Sin, 30, 128, 10.1016/j.dsi.2012.08.004 Festa, 2011, Copper: an essential metal in biology, Curr Biol, 21, R877, 10.1016/j.cub.2011.09.040 LoPresti, 2019, Free radical-decellularized tissue promotes enhanced antioxidant and anti-inflammatory macrophage response, Biomaterials, 222, 10.1016/j.biomaterials.2019.119376 Peng, 2021, Construction of heparin-based hydrogel incorporated with Cu5.4O ultrasmall nanozymes for wound healing and inflammation inhibition, Bioact Mater, 6, 3109, 10.1016/j.bioactmat.2021.02.006 Karlsson, 2009, Size-dependent toxicity of metal oxide particles—a comparison between nano- and micrometer size, Toxicol Lett, 188, 112, 10.1016/j.toxlet.2009.03.014 Wongrakpanich, 2016, Size-dependent cytotoxicity of copper oxide nanoparticles in lung epithelial cells, Environ Sci Nano, 3, 365, 10.1039/C5EN00271K Rehman, 2012, The anti-inflammatory effects of platinum nanoparticles on the lipopolysaccharide-induced inflammatory response in RAW 264.7 macrophages, Inflamm Res, 61, 1177, 10.1007/s00011-012-0512-0 Yoshihisa, 2010, Protective effects of platinum nanoparticles against UV-light-induced epidermal inflammation, Exp Dermatol, 19, 1000, 10.1111/j.1600-0625.2010.01128.x Onizawa, 2009, Platinum nanoparticle antioxidants inhibit pulmonary inflammation in mice exposed to cigarette smoke, Pulm Pharmacol Ther, 22, 340, 10.1016/j.pupt.2008.12.015 Takamiya, 2011, Neurological and pathological improvements of cerebral infarction in mice with platinum nanoparticles, J Neurosci Res, 89, 1125, 10.1002/jnr.22622 Adeyemi, 2016, Biochemical and morphological changes in rats exposed to platinum nanoparticles, Comp Clin Pathol, 25, 855, 10.1007/s00580-016-2274-5 Hirst, 2009, Anti-inflammatory properties of cerium oxide nanoparticles, Small, 5, 2848, 10.1002/smll.200901048 Celardo, 2011, Pharmacological potential of cerium oxide nanoparticles, Nanoscale, 3, 1411, 10.1039/c0nr00875c Yokel, 2013, Biodistribution and biopersistence of ceria engineered nanomaterials: size dependence, Nanomedicine, 9, 398, 10.1016/j.nano.2012.08.002 Hirst, 2013, Bio-distribution and in vivo antioxidant effects of cerium oxide nanoparticles in mice, Environ Toxicol, 28, 107, 10.1002/tox.20704 Yokel, 2009, Biodistribution and oxidative stress effects of a systemically-introduced commercial ceria engineered nanomaterial, Nanotoxicology, 3, 234, 10.1080/17435390902974496 Modrzynska, 2018, In vivo-induced size transformation of cerium oxide nanoparticles in both lung and liver does not affect long-term hepatic accumulation following pulmonary exposure, PLoS One, 13, 10.1371/journal.pone.0202477 Yousefi, 2018, Pharmacokinetics and biodistribution of pegylated methotrexate after IV administration to mice, Iran J Pharm Res (IJPR), 17, 111 Guo, 2018, Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies, Bone Res, 6, 15, 10.1038/s41413-018-0016-9 Cheng, 2013, Cerium oxide nanoparticles induce cytotoxicity in human hepatoma SMMC-7721 cells via oxidative stress and the activation of MAPK signaling pathways, Toxicol Vitro, 27, 1082, 10.1016/j.tiv.2013.02.005 Wang, 2015, Effects of nano-CeO2 with different nanocrystal morphologies on cytotoxicity in HepG2 cells, Int J Environ Res Publ Health, 12, 10806, 10.3390/ijerph120910806 Forest, 2017, Impact of cerium oxide nanoparticles shape on their in vitro cellular toxicity, Toxicol Vitro, 38, 136, 10.1016/j.tiv.2016.09.022 Hijaz, 2016, Folic acid tagged nanoceria as a novel therapeutic agent in ovarian cancer, BMC Cancer, 16, 220, 10.1186/s12885-016-2206-4 Tripathy, 2018, Metal oxide modified ZnO nanomaterials for biosensor applications, Nano Converg, 5, 27, 10.1186/s40580-018-0159-9 Martínez-Carmona, 2018, ZnO nanostructures for drug delivery and theranostic applications, Nanomaterials (Basel), 8, 268, 10.3390/nano8040268 Kim, 2014, Zinc oxide nanoparticles, a novel candidate for the treatment of allergic inflammatory diseases, Eur J Pharmacol, 738, 31, 10.1016/j.ejphar.2014.05.030 Nagajyothi, 2015, Antioxidant and anti-inflammatory activities of zinc oxide nanoparticles synthesized using Polygala tenuifolia root extract, J Photochem Photobiol, B, 146, 10, 10.1016/j.jphotobiol.2015.02.008 Choi, 2015, Toxicity of zinc oxide nanoparticles in rats treated by two different routes: single intravenous injection and single oral administration, J Toxicol Environ Health A, 78, 226, 10.1080/15287394.2014.949949 Jo, 2013, Exposure to zinc oxide nanoparticles affects reproductive development and biodistribution in offspring rats, J Toxicol Sci, 38, 525, 10.2131/jts.38.525 Cho, 2013, Comparative absorption, distribution, and excretion of titanium dioxide and zinc oxide nanoparticles after repeated oral administration, Part Fibre Toxicol, 10, 9, 10.1186/1743-8977-10-9 Li, 2012, Organ biodistribution, clearance, and genotoxicity of orally administered zinc oxide nanoparticles in mice, Nanotoxicology, 6, 746, 10.3109/17435390.2011.620717 Paek, 2013, Modulation of the pharmacokinetics of zinc oxide nanoparticles and their fates in vivo, Nanoscale, 5, 11416, 10.1039/c3nr02140h Baek, 2012, Pharmacokinetics, tissue distribution, and excretion of zinc oxide nanoparticles, Int J Nanomed, 7, 3081 Wang, 2016, Effects of long-term exposure to zinc oxide nanoparticles on development, zinc metabolism and biodistribution of minerals (Zn, Fe, Cu, Mn) in mice, PLoS One, 11 Kang, 2013, In vitro toxicity of different-sized ZnO nanoparticles in Caco-2 cells, Nanoscale Res Lett, 8, 496, 10.1186/1556-276X-8-496 Baek, 2011, Factors influencing the cytotoxicity of zinc oxide nanoparticles: particle size and surface charge, J Phys Conf Ser, 304, 10.1088/1742-6596/304/1/012044 Wingett, 2016, A role of ZnO nanoparticle electrostatic properties in cancer cell cytotoxicity, Nanotechnol Sci Appl, 9, 29, 10.2147/NSA.S99747 Jafari, 2020, Biomedical applications of TiO2 nanostructures: recent advances, Int J Nanomed, 15, 3447, 10.2147/IJN.S249441 Seisenbaeva, 2017, Dispersion of TiO2 nanoparticles improves burn wound healing and tissue regeneration through specific interaction with blood serum proteins, Sci Rep, 7, 10.1038/s41598-017-15792-w Liu, 2016, Bioactive SiO2@Ru nanoparticles for osteogenic differentiation of mesenchymal stem cells via activation of Akt signaling pathways, J Mater Chem B, 4, 4389, 10.1039/C5TB01898F Lu, 2021, A dual-targeting ruthenium nanodrug that inhibits primary tumor growth and lung metastasis via the PARP/ATM pathway, J Nanobiotechnol, 19, 115, 10.1186/s12951-021-00799-3 Hoshyar, 2016, The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction, Nanomedicine (Lond), 11, 673, 10.2217/nnm.16.5 Srinivasarao, 2017, Ligand-targeted drug delivery, Chem Rev, 117, 12133, 10.1021/acs.chemrev.7b00013 Wang, 2017, Peptide and antibody ligands for renal targeting: nanomedicine strategies for kidney disease, Biomater Sci, 5, 1450, 10.1039/C7BM00271H Cheng, 2020, Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing, Nat Nanotechnol, 15, 313, 10.1038/s41565-020-0669-6 Yin, 2019, Intrapulmonary cellular-level distribution of inhaled nanoparticles with defined functional groups and its correlations with protein corona and inflammatory response, ACS Nano, 13, 14048, 10.1021/acsnano.9b06424 Hansson, 2006, The immune response in atherosclerosis: a double-edged sword, Nat Rev Immunol, 6, 508, 10.1038/nri1882 Zhang, 2019, Promoting the delivery of nanoparticles to atherosclerotic plaques by DNA coating, ACS Appl Mater Interfaces, 11, 13888, 10.1021/acsami.8b17928 Feng, 2019, Immunomodulatory nanosystems, Adv Sci (Weinh), 6 Martin, 2011, Metal ion toxicity Ibrahim, 2006, Heavy metal poisoning: clinical presentations and pathophysiology, Clin Lab Med, 26, 67, 10.1016/j.cll.2006.02.003 Sigler, 1974, Gold salts in the treatment of rheumatoid arthritis, Ann Intern Med, 80, 21, 10.7326/0003-4819-80-1-21 Beckett, 1982, Chrysiasis resulting from gold therapy in rheumatoid arthritis: identification of gold by X-ray microanalysis, Mayo Clin Proc, 57, 773 Desoize, 2002, Particular aspects of platinum compounds used at present in cancer treatment, Crit Rev Oncol Hematol, 42, 317, 10.1016/S1040-8428(01)00219-0 Gaetke, 2003, Copper toxicity, oxidative stress, and antioxidant nutrients, Toxicology, 189, 147, 10.1016/S0300-483X(03)00159-8 Ahuja, 2015, Copper mediated neurological disorder: visions into amyotrophic lateral sclerosis, Alzheimer and Menkes disease, J Trace Elem Med Biol, 29, 11, 10.1016/j.jtemb.2014.05.003 Fosmire, 1990, Zinc toxicity, Am J Clin Nutr, 51, 225, 10.1093/ajcn/51.2.225 Zheng, 2021, Challenges and opportunities of nanomedicines in clinical translation, BIO Integration, 2, 57, 10.15212/bioi-2021-0016 Boehncke, 2007, Animal models of psoriasis, Clin Dermatol, 25, 596, 10.1016/j.clindermatol.2007.08.014 Male, 2008, Synthesis and stability of fluorescent gold nanoparticles by sodium borohydride in the presence of mono-6-deoxy-6-pyridinium-β-cyclodextrin chloride, J Phys Chem C, 112, 443, 10.1021/jp7099515 Benítez, 2009, The use of ultrafiltration and nanofiltration membranes for the purification of cork processing wastewater, J Hazard Mater, 162, 1438, 10.1016/j.jhazmat.2008.06.036 Jung, 2015, Removal of humic and tannic acids by adsorption–coagulation combined systems with activated biochar, J Hazard Mater, 300, 808, 10.1016/j.jhazmat.2015.08.025 Foulkes, 2020, The regulation of nanomaterials and nanomedicines for clinical application: current and future perspectives, Biomater Sci, 8, 4653, 10.1039/D0BM00558D 2022