1
|
Kulich P, Marvanová S, Skoupý R, Škorič M, Vysloužil J, Šerý O, Mikuška P, Alexa L, Coufalík P, Křůmal K, Moravec P, Večeřa Z, Machala M. Subchronic Inhalation of TiO 2 Nanoparticles Leads to Deposition in the Lung and Alterations in Erythrocyte Morphology in Mice. J Appl Toxicol 2025; 45:1004-1018. [PMID: 39933250 PMCID: PMC12061548 DOI: 10.1002/jat.4759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
TiO2 nanoparticles (NPs) are extensively used in various applications, highlighting the importance of ongoing research into their effects. This work belongs among rare whole-body inhalation studies investigating the effects of TiO2 NPs on mice. Unlike previous studies, the concentration of TiO2 NPs in the inhalation chamber (130.8 μg/m3) was significantly lower. This 11-week study on mice confirmed in vivo the presence of TiO2 NPs in lung macrophages and type II pneumocytes including their intracellular localization by using the electron microscopy and the state-of-the-art methods detecting NPs' chemical identity/crystal structure, such as the energy-dispersed X-ray spectroscopy (EDX), cathodoluminescence (CL), and detailed diffraction pattern analysis using powder nanobeam diffraction (PNBD). For the first time in inhalation study in vivo, the alterations in erythrocyte morphology with evidence of echinocytes and stomatocytes, accompanied by iron accumulation in spleen, liver, and kidney, are reported following NP's exposure. Together with the histopathological evidence of hyperaemia in the spleen and kidney, and haemosiderin presence in the spleen, the finding of NPs containing iron might suggest the increased decomposition of damaged erythrocytes. The detection of TiO2 NPs on erythrocytes through CL analysis confirmed their potential systemic availability. On the contrary, TiO2 NPs were not confirmed in other organs (spleen, liver, and kidney); Ti was detected only in the kidney near the detection limit.
Collapse
Affiliation(s)
- Pavel Kulich
- Department of Pharmacology and ToxicologyVeterinary Research InstituteBrnoCzech Republic
| | - Soňa Marvanová
- Department of Pharmacology and ToxicologyVeterinary Research InstituteBrnoCzech Republic
| | - Radim Skoupý
- Institute of Scientific InstrumentsCzech Academy of SciencesBrnoCzech Republic
- Department of BionanoscienceDelft University of TechnologyDelftThe Netherlands
- Kavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| | - Miša Škorič
- Department of Pathological Morphology and Parasitology, Faculty of Veterinary MedicineUniversity of Veterinary SciencesBrnoCzech Republic
| | - Jan Vysloužil
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Omar Šerý
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and GeneticsCzech Academy of SciencesBrnoCzech Republic
| | - Pavel Mikuška
- Department of Environmental Analytical Chemistry, Institute of Analytical ChemistryCzech Academy of SciencesBrnoCzech Republic
| | - Lukáš Alexa
- Department of Environmental Analytical Chemistry, Institute of Analytical ChemistryCzech Academy of SciencesBrnoCzech Republic
| | - Pavel Coufalík
- Department of Environmental Analytical Chemistry, Institute of Analytical ChemistryCzech Academy of SciencesBrnoCzech Republic
| | - Kamil Křůmal
- Department of Environmental Analytical Chemistry, Institute of Analytical ChemistryCzech Academy of SciencesBrnoCzech Republic
| | - Pavel Moravec
- Department of Environmental Engineering, Institute of Chemical Process FundamentalsCzech Academy of SciencesPragueCzech Republic
| | - Zbyněk Večeřa
- Department of Environmental Analytical Chemistry, Institute of Analytical ChemistryCzech Academy of SciencesBrnoCzech Republic
| | - Miroslav Machala
- Department of Pharmacology and ToxicologyVeterinary Research InstituteBrnoCzech Republic
| |
Collapse
|
2
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
3
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2025; 13:2164-2178. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
4
|
Russo RC, Togbe D, Couillin I, Segueni N, Han L, Quesniaux VFJ, Stoeger T, Ryffel B. Ozone-induced lung injury and inflammation: Pathways and therapeutic targets for pulmonary diseases caused by air pollutants. ENVIRONMENT INTERNATIONAL 2025; 198:109391. [PMID: 40121788 DOI: 10.1016/j.envint.2025.109391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Exposure to ambient Ozone (O3) air pollution directly causes by its oxidative properties, respiratory epithelial cell injury, and cell death, which promote inflammation and hyperreactivity, posing a significant public health concern. Recent clinical and experimental studies have made strides in elucidating the mechanisms underlying O3-induced epithelial cell injury, inflammation, and airway hyperreactivity, which are discussed herein. The current data suggest that O3-induced oxidative stress is a central event-inducing oxeiptotic cell death pathway. O3-induced epithelial barrier damage and cell death, triggering the release of alarmins and damage-associated molecular patterns (DAMPs), with subsequent endogenous activation of Toll-like receptors (TLRs), DNA sensing pathways, and inflammasomes, activating interleukin-1-Myd88 inflammatory pathway with the production of a range of chemokines and cytokines. This cascade orchestrates lung tissue-resident cell activation in response to O3 in leukocyte and non-leukocyte populations, driving sterile innate immune response. Chronic inflammatory response to O3, by repeated exposures, supports a mixed phenotype combining asthma and emphysema, in which their exacerbation by other particulate pollutants potentially culminates in respiratory failure. We use data from lung single-cell transcriptomics to map genes of O3-damage sensing and signaling pathways to lung cells and thereby highlight potential hotspots of O3 responses. Deeper insights into these pathological pathways might be helpful for the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Dieudonnée Togbe
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Isabelle Couillin
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | | | - Lianyong Han
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Valérie F J Quesniaux
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France; ArtImmune SAS, 13 Avenue Buffon, Orleans, France.
| |
Collapse
|
5
|
Aschner M, Skalny AV, Martins AC, Tizabi Y, Zaitseva IP, Santamaria A, Lu R, Gluhcheva YY, Tinkov AA. The role of NLRP3 inflammasome activation in proinflammatory and cytotoxic effects of metal nanoparticles. Arch Toxicol 2025; 99:1287-1314. [PMID: 39960653 DOI: 10.1007/s00204-025-03972-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/27/2025] [Indexed: 04/04/2025]
Abstract
Exposure to metal nanoparticles (NPs) is known to induce inflammatory responses in various tissues, thus limiting their therapeutic potential. NOD-like receptor protein 3 (NLRP3) inflammasome activation is an essential component of innate immunity playing a significant role in inflammation and development of inflammatory diseases. Therefore, the objective of the present review was to summarize data on the role of NLRP3 inflammasome in proinflammatory effects induced by metal NPs, and to discuss the underlying molecular mechanisms, including its dependence on the physical and chemical properties of metal NPs. Titanium, zinc, silver, aluminum, iron, cobalt, nickel, vanadium, and tungsten nanoparticles, as well as metal-based quantum dots have all been shown to induce NLRP3 inflammasome activation in vitro in macrophages and monocytes, dendritic cells, keratinocytes, hepatocytes, enterocytes, microglia, astrocytes, lung epithelial cells, endotheliocytes, as well as certain types of cancer cells. In vivo studies confirmed the role of NLRP3 pathway activation in development of colitis, pulmonary inflammation, liver damage, osteolysis, and neuroinflammation induced by various metal nanoparticles. Briefly, particle endocytosis with subsequent lysosomal damage, induction of ROS formation, K+ efflux, increased intracellular Ca2+ levels, and NF-κB pathway activation results in NLRP3 inflammasome complex assembly, caspase-1 activation, and cleavage of pro-IL-1β and pro-IL-18 to mature proinflammatory cytokines, while gasdermin D cleavage induces pyroptotic cell death. Moreover, small-sized and rod-shaped metal NPs exert a more profound stimulatory effect on NLRP3 inflammasome activation, but contrary findings have also been reported. Taken together, it is concluded that NLRP3 inflammasome may mediate both adverse proinflammatory effects of metal nanoparticles, as well as their beneficial effect when used as antitumor agents.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Irina P Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control and Department of Physical Education, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yordanka Y Gluhcheva
- Institute of Experimental Morphology, Pathology and Anthropology With Museum, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str., Bl. 25, 1113, Sofia, Bulgaria
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
- Laboratory of Ecobiomonitoring and Quality Control and Department of Physical Education, Yaroslavl State University, Yaroslavl, 150003, Russia
| |
Collapse
|
6
|
Saosamniang P, Matsumura K, Okajima MK, Kaneko T. Directing mineralization of ZnO nanoparticles in cyanobacterial liquid crystalline polysaccharides for cancer therapies. Int J Biol Macromol 2025; 304:140716. [PMID: 39920940 DOI: 10.1016/j.ijbiomac.2025.140716] [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: 08/29/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Effective cancer therapy faces significant challenges, including non-selective toxicity, limited structural stability, inconsistent nanoparticle (NP) morphology, and instability under varying biological conditions. These issues hindering targeted delivery and therapeutic efficacy. Previous approaches using polysaccharide-based nanomaterials have shown promise; however, problems such as inconsistent NP sizes and shapes, poor mechanical stability, and limited pH resilience restrict their clinical potential. This study hypothesized that sacran, a cyanobacterial liquid crystalline (LC) polysaccharide, can stabilize ZnO NPs, allowing for controlled mineralization, enhanced stability, and selective cytotoxicity. We developed ZnO nanocomposite xerogels in an LC sacran matrix, yielding block-like ZnO NPs (25-70 nm) with high surface-area-to-volume ratios that improve cellular uptake in tumor environments. Incorporating these NPs into chemically crosslinked sacran matrices resulted in a 3-fold increase in mechanical strength and a 10-fold improvement in swelling capacity compared to physically crosslinked systems. Additionally, the sacran-ZnO nanocomposites demonstrated robust stability under various pH conditions, indicating their resilience in diverse biological environments. Cytotoxicity assays revealed that higher concentrations of ZnO NP selectively increased toxicity toward human lung cancer cells (A549), with less impact on human dermal fibroblasts (HDFa). Moreover, HDFa successfully attached to and proliferated on the smooth surfaces of the xerogels, emphasizing their compatibility with normal cells. This highlights the potential of sacran-ZnO nanocomposite xerogels as cancer-selective therapeutic materials, offering stability and effectiveness even under varying biological conditions, while addressing key challenges associated with earlier NP-based therapies.
Collapse
Affiliation(s)
- Pruetsakorn Saosamniang
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kazuaki Matsumura
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Maiko K Okajima
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Tatsuo Kaneko
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
7
|
Li Y, Ren S, Zhou S. Advances in sepsis research: Insights into signaling pathways, organ failure, and emerging intervention strategies. Exp Mol Pathol 2025; 142:104963. [PMID: 40139086 DOI: 10.1016/j.yexmp.2025.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Sepsis is a complex syndrome resulting from an aberrant host response to infection. A hallmark of sepsis is the failure of the immune system to restore balance, characterized by hyperinflammation or immunosuppression. However, the net effect of immune system imbalance and the clinical manifestations are highly heterogeneous among patients. In recent years, research interest has shifted from focusing on the pathogenicity of microorganisms to the molecular mechanisms of host responses which is also associated with biomarkers that can help early diagnose sepsis and guide treatment decisions. Despite significant advancements in medical science, sepsis remains a major challenge in healthcare, contributing to substantial morbidity and mortality worldwide. Further research is needed to improve our understanding of this condition and develop novel therapies to improve outcomes for patients with sepsis. This review explores the related signal pathways of sepsis and underscores recent advancements in understanding its mechanisms. Exploration of diverse biomarkers and the emerging concept of sepsis endotypes offer promising avenues for precision therapy in the future.
Collapse
Affiliation(s)
- Yehua Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China.
| | - Siying Ren
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Shen'ao Zhou
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
8
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [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: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
9
|
Sung YY, Yang WK, Kim JH, Shin D, Son SJ, Kim SH. Reliea® combination of Codonopsis lanceolata and Chaenomeles sinensis extract alleviates airway inflammation on particulate matter 10 plus diesel exhaust particles (PM 10D) ‑induced respiratory disease mouse model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117538. [PMID: 39674023 DOI: 10.1016/j.ecoenv.2024.117538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Particulate matter (PM, diameter < 10 μm) and Diesel exhaust particles (DEP) exposure can cause severe respiratory disorders. This investigation explored the protective effects of Reliea® (RelA), combination of Codonopsis lanceolata and Chaenomeles sinensis extract, against airway inflammation related to PM10D exposure. RelA treatment suppressed reactive oxygen species, nitric oxide release, cytokine expression (IL-6, IL-1β, iNOS, CXCL-2, MCP-1, and TNF-α), and the related inflammatory mechanisms in PM10-induced alveolar macrophage cells. BALB/c mice were injected with PM10D via intranasal trachea three times over a period of 12 days and RelA were orally dispensed for 12 days. RelA inhibited infiltrating neutrophils, total number of immunocytes in lung and bronchoalveolar lavage fluid (BALF). RelA decreased the expression of interleukin (IL)-17, chemokine (C-X-C motif) ligand (CXCL)-1, thymus and activation-regulated chemokine, macrophage inflammatory protein-2, IL-1α, TNF-α, mucin 5AC, cyclooxygenase-2, and transient receptor potential cation channel subfamily A or V member 1 in BALF and lung, and inhibited IL-1α and macrophage marker F4/80 localization in lung of PM10D-induced mice. RelA treatment decreased serum symmetric dimethyl arginine levels. RelA restored histopathological damage via inhibition of NF-κB and MAPK pathways in the trachea and lung. Lancemaside A and protocatechuic acid as major active compounds of RelA was identified. In addition, RelA showed better expectoration through increased phenol red secretion. These results indicate that Reliea® combination of C. lanceolata and C. sinensis extract might be effective in prevention and treatment of airway inflammation and respiratory diseases.
Collapse
Affiliation(s)
- Yoon-Young Sung
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Won-Kyung Yang
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, Republic of Korea.
| | - Jong Hoon Kim
- Nongshim, R&D Center, 112 (Shindaebang-Dong), Yeouidaebang-Ro, Dongjak-Gu, Seoul, Republic of Korea.
| | - Dongseok Shin
- Nongshim, R&D Center, 112 (Shindaebang-Dong), Yeouidaebang-Ro, Dongjak-Gu, Seoul, Republic of Korea.
| | - Seok June Son
- Nongshim, R&D Center, 112 (Shindaebang-Dong), Yeouidaebang-Ro, Dongjak-Gu, Seoul, Republic of Korea.
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, Republic of Korea.
| |
Collapse
|
10
|
Cao J, Yang Y, Liu X, Huang Y, Xie Q, Kadushkin A, Nedelko M, Wu D, Aquilina NJ, Li X, Cai X, Li R. Deciphering key nano-bio interface descriptors to predict nanoparticle-induced lung fibrosis. Part Fibre Toxicol 2025; 22:1. [PMID: 39810232 PMCID: PMC11731361 DOI: 10.1186/s12989-024-00616-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The advancement of nanotechnology underscores the imperative need for establishing in silico predictive models to assess safety, particularly in the context of chronic respiratory afflictions such as lung fibrosis, a pathogenic transformation that is irreversible. While the compilation of predictive descriptors is pivotal for in silico model development, key features specifically tailored for predicting lung fibrosis remain elusive. This study aimed to uncover the essential predictive descriptors governing nanoparticle-induced pulmonary fibrosis. METHODS We conducted a comprehensive analysis of the trajectory of metal oxide nanoparticles (MeONPs) within pulmonary systems. Two biological media (simulated lung fluid and phagolysosomal simulated fluid) and two cell lines (macrophages and epithelial cells) were meticulously chosen to scrutinize MeONP behaviors. Their interactions with MeONPs, also referred to as nano-bio interactions, can lead to alterations in the properties of the MeONPs as well as specific cellular responses. Physicochemical properties of MeONPs were assessed in biological media. The impact of MeONPs on cell membranes, lysosomes, mitochondria, and cytoplasmic components was evaluated using fluorescent probes, colorimetric enzyme substrates, and ELISA. The fibrogenic potential of MeONPs in mouse lungs was assessed by examining collagen deposition and growth factor release. Random forest classification was employed for analyzing in chemico, in vitro and in vivo data to identify predictive descriptors. RESULTS The nano-bio interactions induced diverse changes in the 4 characteristics of MeONPs and had variable effects on the 14 cellular functions, which were quantitatively evaluated in chemico and in vitro. Among these 18 quantitative features, seven features were found to play key roles in predicting the pro-fibrogenic potential of MeONPs. Notably, IL-1β was identified as the most important feature, contributing 27.8% to the model's prediction. Mitochondrial activity (specifically NADH levels) in macrophages followed closely with a contribution of 17.6%. The remaining five key features include TGF-β1 release and NADH levels in epithelial cells, dissolution in lysosomal simulated fluids, zeta potential, and the hydrodynamic size of MeONPs. CONCLUSIONS The pro-fibrogenic potential of MeONPs can be predicted by combination of key features at nano-bio interfaces, simulating their behavior and interactions within the lung environment. Among the 18 quantitative features, a combination of seven in chemico and in vitro descriptors could be leveraged to predict lung fibrosis in animals. Our findings offer crucial insights for developing in silico predictive models for nano-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Jiayu Cao
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yuhui Yang
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xi Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yang Huang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Qianqian Xie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Aliaksei Kadushkin
- Department of Biological Chemistry, Belarusian State Medical University, Minsk, 220089, Belarus
| | - Mikhail Nedelko
- B.I. Stepanov Institute of Physics of National Academy of Sciences of Belarus, 68Nezalezhnasti Ave, Minsk, 220072, Belarus
| | - Di Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Noel J Aquilina
- Department of Chemistry, University of Malta, Msida, 2080, MSD, Malta
| | - Xuehua Li
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Xiaoming Cai
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China.
- CEET, Nanotechnology Centre, VSB-Technical University of Ostrava, 17 listopadu, Ostrava, 2172-15, 70800, Czech Republic.
| |
Collapse
|
11
|
Gudima A, Hesselbarth D, Li G, Riabov V, Michel J, Liu Q, Schmuttermaier C, Jiao Z, Sticht C, Jawhar A, Obertacke U, Klüter H, Vrana NE, Kzhyshkowska J. Titanium induces proinflammatory and tissue-destructive responses in primary human macrophages. J Leukoc Biol 2024; 116:706-725. [PMID: 38512961 DOI: 10.1093/jleuko/qiae072] [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: 01/17/2023] [Revised: 02/06/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Implants and medical devices are efficient and practical therapeutic solutions for a multitude of pathologies. Titanium and titanium alloys are used in orthopedics, dentistry, and cardiology. Despite very good mechanical properties and corrosion resistance, titanium implants can fail due to inflammatory or tissue degradation-related complications. Macrophages are major immune cells that control acceptance of failure of the implant. In this study, for the first time, we have performed a systematic analysis of the response of differentially activated human macrophages, M(Control), M(IFNγ), and M(IL-4), to the polished and porous titanium surfaces in order to identify the detrimental effect of titanium leading to the tissue destruction and chronic inflammation. Transcriptome analysis revealed that the highest number of differences between titanium and control settings are found in M(IL-4) that model healing type of macrophages. Real-time quantitative polymerase chain reaction analysis confirmed that both polished and porous titanium affected expression of cytokines, chitinases/chitinase-like proteins, and matrix metalloproteinases (MMPs). Titanium-induced release and activation of MMP7 by macrophages was enhanced by fibroblasts in both juxtacrine and paracrine cell interaction models. Production of titanium-induced MMPs and cytokines associated with chronic inflammation was independent of the presence of Staphylococcus aureus. MMP7, one of the most pronounced tissue-destroying factors, and chitinase-like protein YKL-40 were expressed in CD68+ macrophages in peri-implant tissues of patients with orthopedic implants. In summary, we demonstrated that titanium induces proinflammatory and tissue-destructing responses mainly in healing macrophages, and the detrimental effects of titanium surfaces on implant-adjacent macrophages are independent on the bacterial contamination.
Collapse
Affiliation(s)
- Alexandru Gudima
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - David Hesselbarth
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinic for Cardiology and Angiology, University Heart Centre Freiburg-Bad Krozingen, Freiburg, Germany
| | - Guanhao Li
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Vladimir Riabov
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Julia Michel
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Quan Liu
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christina Schmuttermaier
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Zhen Jiao
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Ahmed Jawhar
- Department of Orthopaedics and Trauma Surgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Udo Obertacke
- Department of Orthopaedics and Trauma Surgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Harald Klüter
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Nihal Engin Vrana
- SPARTHA Medical, Strasbourg, France
- Department of Biomaterials and Bioengineering, UMR_S1121, Biomaterials and Bioengineering, INSERM and University of Strasburg, Strasbourg, France
| | - Julia Kzhyshkowska
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
12
|
Rahman L, Williams A, Wu D, Halappanavar S. Polyethylene Terephthalate Microplastics Generated from Disposable Water Bottles Induce Interferon Signaling Pathways in Mouse Lung Epithelial Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1287. [PMID: 39120391 PMCID: PMC11314056 DOI: 10.3390/nano14151287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Microplastics (MPs) are present in ambient air in a respirable size fraction; however, their potential impact on human health via inhalation routes is not well documented. In the present study, methods for a lab-scale generation of MPs from regularly used and littered plastic articles were optimized. The toxicity of 11 different types of MPs, both commercially purchased and in-lab prepared MPs, was investigated in lung epithelial cells using cell viability, immune and inflammatory response, and genotoxicity endpoints. The underlying mechanisms were identified by microarray analysis. Although laborious, the laboratory-scale methods generated a sufficient quantity of well characterized MPs for toxicity testing. Of the 11 MPs tested, the small sized polyethylene terephthalate (PETE) MPs prepared from disposable water bottles induced the maximum toxicity. Specifically, the smaller size PETE MPs induced a robust activation of the interferon signaling pathway, implying that PETE MPs are perceived by cells by similar mechanisms as those employed to recognize pathogens. The PETE MPs of heterogenous size and shapes induced cell injury, triggering cell death, inflammatory cascade, and DNA damage, hallmark in vitro events indicative of potential in vivo tissue injury. The study establishes toxicity of specific types of plastic materials in micron and nano size.
Collapse
Affiliation(s)
- Luna Rahman
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada; (L.R.); (A.W.); (D.W.)
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada; (L.R.); (A.W.); (D.W.)
| | - Dongmei Wu
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada; (L.R.); (A.W.); (D.W.)
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada; (L.R.); (A.W.); (D.W.)
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
13
|
Fadeel B, Keller AA. Nanosafety: a Perspective on Nano-Bio Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310540. [PMID: 38597766 DOI: 10.1002/smll.202310540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Engineered nanomaterials offer numerous benefits to society ranging from environmental remediation to biomedical applications such as drug or vaccine delivery as well as clean and cost-effective energy production and storage, and the promise of a more sustainable way of life. However, as nanomaterials of increasing sophistication enter the market, close attention to potential adverse effects on human health and the environment is needed. Here a critical perspective on nanotoxicological research is provided; the authors argue that it is time to leverage the knowledge regarding the biological interactions of nanomaterials to achieve a more comprehensive understanding of the human health and environmental impacts of these materials. Moreover, it is posited that nanomaterials behave like biological entities and that they should be regulated as such.
Collapse
Affiliation(s)
- Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Arturo A Keller
- Bren School of Environmental Science & Management, University of California Santa Barbara, California, CA, 93106, USA
| |
Collapse
|
14
|
Couderc LJ, Fleury-Feith J, Longchampt E, Wagner I, Rivaud E, Brun AL, Bernaudin JF, Catherinot E, Kahn JE. Titanium dental implants-related acute pneumonitis. Respir Med Res 2024; 85:101104. [PMID: 38657296 DOI: 10.1016/j.resmer.2024.101104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/26/2024]
Affiliation(s)
- Louis-Jean Couderc
- Service de Pneumologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France; Université Versailles- Saint Quentin, Paris-Saclay UMR 0892 Virologie et Immunologie Moléculaire, 11 rue Guillaume Lenoir, 92150, Suresnes, France.
| | - Jocelyne Fleury-Feith
- Unité de Microscopie Electronique, Hôpital Intercommunal, 40 Av de Verdun, 94010, Créteil, France; Service Parisien de Santé Environnementale, Laboratoire Amiante, Fibres et Particules, Ville de Paris, 11 rue Georges Eastman, 75013, Paris, France; Faculté de Médecine Paris-Sorbonne, 91-105 Bd de l'Hôpital, 75013, Paris, France
| | - Elisabeth Longchampt
- Service d'Anatomo-Pathologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France
| | - Isabelle Wagner
- Service d'Oto-Rhino-Laryngologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France
| | - Elisabeth Rivaud
- Service de Pneumologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France
| | - Anne-Laure Brun
- Service de Radiologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France
| | - Jean-François Bernaudin
- Faculté de Médecine Paris-Sorbonne, 91-105 Bd de l'Hôpital, 75013, Paris, France; INSERM UMR 1272 Universite Sorbonne Paris-Nord, SMBH, 1 rue de Chablis, 93017, Bobigny, France
| | - Emilie Catherinot
- Service de Pneumologie, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France
| | - Jean-Emmanuel Kahn
- Service de Médecine Interne, Hôpital Foch, 40 rue Worth, 92150, Suresnes, France; Université Versailles-Saint Quentin Paris-Saclay, Faculté des Sciences de la Vie Simone Veil, 2 Av de la source de la Bièvre, 78180, Montigny-le-Bretonneux, France
| |
Collapse
|
15
|
Kühnel D, Krug HF, Steinbach C, Nau K. The DaNa projects: public communication of (nano)material safety data-from conspiracy theories to study quality. FRONTIERS IN TOXICOLOGY 2024; 6:1382458. [PMID: 38863790 PMCID: PMC11165057 DOI: 10.3389/ftox.2024.1382458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/02/2024] [Indexed: 06/13/2024] Open
Abstract
In this perspective, the authors give their view on the developments and experiences on communicating on (nano)materials safety. We would like to share our experiences with the scientific community in order to make them useful for future communication activities. We present the long-term work of the science communication projects DaNa, DaNa2.0 and DaNa4.0, running from 2009 to 2023. Starting in the early 2000s with the beginnings of nanotechnology research, communication on the safety of nanomaterials with the public was still very new and faced the projects with many challenges. Today, science communication is indispensable for the dissemination of scientific findings and a fact-based approach like the DaNa "Knowledge Base Materials" creates a trustworthy dialogue with the public. This long-term project series has made a significant contribution to communication on the safety of nanomaterials, perhaps even the largest among publicly funded project series worldwide.
Collapse
Affiliation(s)
- Dana Kühnel
- Helmholtz Centre for Environmental Research (UFZ), Department Ecotoxicology (ETOX), Leipzig, Germany
| | | | - Christoph Steinbach
- Society for Chemical Engineering and Biotechnology (DECHEMA), Frankfurt am Main, Germany
| | - Katja Nau
- Karlsruhe Institute of Technology (KIT), Institute for Automation and Applied Informatics (IAI), Karlsruhe, Germany
| |
Collapse
|
16
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Hasegawa T, Noguchi S, Nakashima M, Miyai M, Goto M, Matsumoto Y, Torii S, Honda S, Shimizu S. Alternative autophagy dampens UVB-induced NLRP3 inflammasome activation in human keratinocytes. J Biol Chem 2024; 300:107173. [PMID: 38499149 PMCID: PMC11002869 DOI: 10.1016/j.jbc.2024.107173] [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: 09/01/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
Sunlight exposure results in an inflammatory reaction of the skin commonly known as sunburn, which increases skin cancer risk. In particular, the ultraviolet B (UVB) component of sunlight induces inflammasome activation in keratinocytes to instigate the cutaneous inflammatory responses. Here, we explore the intracellular machinery that maintains skin homeostasis by suppressing UVB-induced inflammasome activation in human keratinocytes. We found that pharmacological inhibition of autophagy promoted UVB-induced NLRP3 inflammasome activation. Unexpectedly, however, gene silencing of Atg5 or Atg7, which are critical for conventional autophagy, had no effect, whereas gene silencing of Beclin1, which is essential not only for conventional autophagy but also for Atg5/Atg7-independent alternative autophagy, promoted UVB-induced inflammasome activation, indicating an involvement of alternative autophagy. We found that damaged mitochondria were highly accumulated in UVB-irradiated keratinocytes when alternative autophagy was inhibited, and they appear to be recognized by NLRP3. Overall, our findings indicate that alternative autophagy, rather than conventional autophagy, suppresses UVB-induced NLRP3 inflammasome activation through the clearance of damaged mitochondria in human keratinocytes and illustrate a previously unknown involvement of alternative autophagy in inflammation. Alternative autophagy may be a new therapeutic target for sunburn and associated cutaneous disorders.
Collapse
Affiliation(s)
| | - Saori Noguchi
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | - Makiko Goto
- Shiseido Global Innovation Center, Yokohama, Japan
| | | | - Satoru Torii
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
18
|
Rios-Ibarra CP, Salinas-Santander M, Orozco-Nunnelly DA, Bravo-Madrigal J. Nanoparticle‑based antiviral strategies to combat the influenza virus (Review). Biomed Rep 2024; 20:65. [PMID: 38476608 PMCID: PMC10928480 DOI: 10.3892/br.2024.1753] [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: 10/20/2023] [Accepted: 01/17/2024] [Indexed: 03/14/2024] Open
Abstract
The rapid availability of effective antiviral treatments would be beneficial during the early phases of a pandemic, as they could reduce viral loads and control serious infections until antigenic vaccines become widely available. One promising alternative therapy to combat pandemics is nanotechnology, which has the potential to inhibit a wide variety of viruses, including the influenza virus. This review summarizes the recent progress using gold, copper, silver, silicone, zinc and selenium nanoparticles, since these materials have shown remarkable antiviral capacity against influenza A virus.
Collapse
Affiliation(s)
- Clara Patricia Rios-Ibarra
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of The State of Jalisco (CIATEJ), Guadalajara, Jalisco 44270, Mexico
| | - Mauricio Salinas-Santander
- Research Department, School of Medicine Saltillo, Universidad Autonoma de Coahuila, Unidad Saltillo, Coahuila 25000, Mexico
| | | | - Jorge Bravo-Madrigal
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of The State of Jalisco (CIATEJ), Guadalajara, Jalisco 44270, Mexico
| |
Collapse
|
19
|
Wang BJ, Chen YY, Chang HH, Chen RJ, Wang YJ, Lee YH. Zinc oxide nanoparticles exacerbate skin epithelial cell damage by upregulating pro-inflammatory cytokines and exosome secretion in M1 macrophages following UVB irradiation-induced skin injury. Part Fibre Toxicol 2024; 21:9. [PMID: 38419076 PMCID: PMC10900617 DOI: 10.1186/s12989-024-00571-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: 09/06/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Zinc oxide nanoparticles (ZnONPs) are common materials used in skin-related cosmetics and sunscreen products due to their whitening and strong UV light absorption properties. Although the protective effects of ZnONPs against UV light in intact skin have been well demonstrated, the effects of using ZnONPs on damaged or sunburned skin are still unclear. In this study, we aimed to reveal the detailed underlying mechanisms related to keratinocytes and macrophages exposed to UVB and ZnONPs. RESULTS We demonstrated that ZnONPs exacerbated mouse skin damage after UVB exposure, followed by increased transepidermal water loss (TEWL) levels, cell death and epithelial thickness. In addition, ZnONPs could penetrate through the damaged epithelium, gain access to the dermis cells, and lead to severe inflammation by activation of M1 macrophage. Mechanistic studies indicated that co-exposure of keratinocytes to UVB and ZnONPs lysosomal impairment and autophagy dysfunction, which increased cell exosome release. However, these exosomes could be taken up by macrophages, which accelerated M1 macrophage polarization. Furthermore, ZnONPs also induced a lasting inflammatory response in M1 macrophages and affected epithelial cell repair by regulating the autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. CONCLUSIONS Our findings propose a new concept for ZnONP-induced skin toxicity mechanisms and the safety issue of ZnONPs application on vulnerable skin. The process involved an interplay of lysosomal impairment, autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. The current finding is valuable for evaluating the effects of ZnONPs for cosmetics applications.
Collapse
Affiliation(s)
- Bour-Jr Wang
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
- Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, 70403, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Hui-Hsuan Chang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 406040, Taiwan.
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
20
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
21
|
Insua A, Galindo-Moreno P, Miron RJ, Wang HL, Monje A. Emerging factors affecting peri-implant bone metabolism. Periodontol 2000 2024; 94:27-78. [PMID: 37904311 DOI: 10.1111/prd.12532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/05/2023] [Accepted: 09/10/2023] [Indexed: 11/01/2023]
Abstract
Implant dentistry has evolved to the point that standard implant osseointegration is predictable. This is attributed in part to the advancements in material sciences that have led toward improvements in implant surface technology and characteristics. Nonetheless, there remain several cases where implant therapy fails (specifically at early time points), most commonly attributed to factors affecting bone metabolism. Among these patients, smokers are known to have impaired bone metabolism and thus be subject to higher risks of early implant failure and/or late complications related to the stability of the peri-implant bone and mucosal tissues. Notably, however, emerging data have unveiled other critical factors affecting osseointegration, namely, those related to the metabolism of bone tissues. The aim of this review is to shed light on the effects of implant-related factors, like implant surface or titanium particle release; surgical-related factors, like osseodensification or implanted biomaterials; various drugs, like selective serotonin reuptake inhibitors, proton pump inhibitors, anti-hypertensives, nonsteroidal anti-inflammatory medication, and statins, and host-related factors, like smoking, diet, and metabolic syndrome on bone metabolism, and aseptic peri-implant bone loss. Despite the infectious nature of peri-implant biological complications, these factors must be surveyed for the effective prevention and management of peri-implantitis.
Collapse
Affiliation(s)
- Angel Insua
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pablo Galindo-Moreno
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Oral Surgery and Implant Dentistry, University of Granada, Granada, Spain
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Hom-Lay Wang
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alberto Monje
- Department of Periodontology and Oral Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Periodontology, University of Bern, Bern, Switzerland
- Department of Periodontology, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
22
|
Li C, Tang M. The toxicological effects of nano titanium dioxide on target organs and mechanisms of toxicity. J Appl Toxicol 2024; 44:152-164. [PMID: 37655586 DOI: 10.1002/jat.4534] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/04/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Nano-titanium dioxide (TiO2 NPs) is widely used for its extremely high stability, corrosion resistance, and photocatalytic properties and has penetrated into various fields of production and life. Assessing its toxicity to different organs should be a key part of preclinical toxicity assessment of TiO2 NPs, which is relatively incomprehensive yet. Therefore, this review focuses on the toxic effects of TiO2 NPs on various organs in mammals and biological mechanisms from different organs. The commonality of toxic effects on various target organs reflected in tissue structure damage and dysfunction, such as liver damage and dysfunction; pulmonary fibrosis; and renal impairment (including hematuria and nephritis); damage of brain tissue and neurons; alteration of intestinal villi; and weight loss. And effects on the reproductive system are affected by different sexes, including ovarian dysfunction, testicular development damage, and sperm viability reduction. We believe that the toxic mechanisms of TiO2 NPs in target organs have commonalities, such as oxidative stress, inflammatory responses, and organelle damage. However, different target organ toxicities also have their specificities. TiO2 NPs disturb the intestinal flora and cause undesirable changes in feces products. And in spleen are infiltration of neutrophils and lymphadenopathy and eventually immune deficiency. Although the toxic pathways are different, but there may be a close link between the different toxic pathways. In this article, the main manifestations of the toxic effects of titanium dioxide nanoparticles on major mammalian organs are reviewed, in order to provide basic data for their better application from a medical perspective.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Environmental Medicine of Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Meng Tang
- Key Laboratory of Environmental Medicine of Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
23
|
Dong T, Yu C, Yang Z, He G, Wen Y, Roseng LE, Wei X, Jing W, Lin Q, Zhao L, Jiang Z. Nanotoxicity of tungsten trioxide nanosheets containing oxygen vacancy to human umbilical vein endothelial cells. Colloids Surf B Biointerfaces 2024; 234:113742. [PMID: 38271855 DOI: 10.1016/j.colsurfb.2023.113742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/09/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024]
Abstract
Because of the excellent performance in photochemistry, WO3 is increasingly applied in the field of biology and medicine. However, little is known about the mechanism of WO3 cytotoxicity. In this work, WO3 nanosheets with oxygen vacancy are synthesized by solvothermal method, then characterized and added to culture medium of human umbilical vein endothelial cells (HUVECs) with different concentrations. We characterized and analyzed the morphology of nano-WO3 by transmission electron microscopy and calculated the specific data of oxygen vacancy by XPS. It is the first time the effect of WO3-x on cells that WO3-x can cause oxidative stress in HUVEC cells, resulting in DNA damage and thus promoting apoptosis. Transcriptome sequencing is performed on cells treated with low and high concentrations of WO3-x, and a series of key signals affecting cell proliferation and apoptosis are detected in differentially expressed genes, which indicates the research direction of nanotoxicity. The expression levels of key genes are also verified by quantitative PCR after cell treatment with different concentrations of WO3-x. This work fills the gap between the biocompatibility of nano WO3-x materials and molecular cytology and paves the way for investigating the mechanism and risks of oxygen vacancy in cancer therapy.
Collapse
Affiliation(s)
- Tao Dong
- Chongqing Key Laboratory of Micro-Nano Systems and Intelligent Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-NanoSystems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China; Department of Microsystems (IMS), Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Postboks 235, 3603 Kongsberg, Norway.
| | - Chenghui Yu
- Chongqing Key Laboratory of Micro-Nano Systems and Intelligent Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-NanoSystems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China
| | - Zhaochu Yang
- Chongqing Key Laboratory of Micro-Nano Systems and Intelligent Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-NanoSystems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China.
| | - Guozhen He
- Chongqing Key Laboratory of Micro-Nano Systems and Intelligent Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-NanoSystems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China; Department of Microsystems (IMS), Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Postboks 235, 3603 Kongsberg, Norway
| | - Yumei Wen
- Department of Instrumentation Science and Engineering, Shanghai Jiao Tong University, Minhang District, Shanghai 200240, China
| | - Lars Eric Roseng
- Department of Microsystems (IMS), Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Postboks 235, 3603 Kongsberg, Norway
| | - Xueyong Wei
- School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Weixuan Jing
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Qijing Lin
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Libo Zhao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhuangde Jiang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
24
|
Ma Q, Lim CS. Molecular Activation of NLRP3 Inflammasome by Particles and Crystals: A Continuing Challenge of Immunology and Toxicology. Annu Rev Pharmacol Toxicol 2024; 64:417-433. [PMID: 37708431 PMCID: PMC10842595 DOI: 10.1146/annurev-pharmtox-031023-125300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Particles and crystals constitute a unique class of toxic agents that humans are constantly exposed to both endogenously and from the environment. Deposition of particulates in the body is associated with a range of diseases and toxicity. The mechanism by which particulates cause disease remains poorly understood due to the lack of mechanistic insights into particle-biological interactions. Recent research has revealed that many particles and crystals activate the NLRP3 inflammasome, an intracellular pattern-recognition receptor. Activated NLRP3 forms a supramolecular complex with an adaptor protein to activate caspase 1, which in turn activates IL-1β and IL-18 to instigate inflammation. Genetic ablation and pharmacological inhibition of the NLRP3 inflammasome dampen inflammatory responses to particulates. Nonetheless, how particulates activate NLRP3 remains a challenging question. From this perspective, we discuss our current understanding of and progress on revealing the function and mode of action of the NLRP3 inflammasome in mediating adaptive and pathologic responses to particulates in health and disease.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia, USA;
| | - Chol Seung Lim
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia, USA;
| |
Collapse
|
25
|
Zhuo LB, Liu YM, Jiang Y, Yan Z. Zinc oxide nanoparticles induce acute lung injury via oxidative stress-mediated mitochondrial damage and NLRP3 inflammasome activation: In vitro and in vivo studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122950. [PMID: 37979646 DOI: 10.1016/j.envpol.2023.122950] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
The widespread application of zinc oxide nanoparticles (ZnO-NPs) brings convenience to our lives while also renders threats to public health and ecological environment. The lung has been recognized as a primary target of ZnO-NPs, however, the detrimental effects and mechanism of ZnO-NPs on the respiratory system have not been thoroughly characterized so far. To investigate the effect of ZnO-NPs on acute lung injury (ALI), Sprague Dawley rats were intratracheally instilled with ZnO-NPs suspension at doses of 1, 2, and 4 mg/kg/day for 3 consecutive days. Our study revealed that ZnO-NPs induced ALI in rats characterized by increased airway resistance, excessive inflammatory response and lung histological damage. In addition, we identified several molecular biomarkers related to the potential mechanism of ZnO-NP-induced ALI, including oxidative stress, mitochondrial damage, and NLRP3 inflammasome activation. The results of in vitro experiments showed that the viability of A549 cells decreased with the increase in ZnO-NPs concentration. Meanwhile, it was also found that ZnO-NP treatment induced the production of ROS, the decrease in mitochondrial membrane potential and activation of NLRP3 inflammasome in A549 cells. Furthermore, to explore the underlying molecular mechanisms of ZnO-NP-induced ALI, N-acetyl-L-cysteine (a ROS scavenger), Cyclosporin A (an inhibitor for mitochondrial depolarization) and Glibenclamide (an inhibitor for NLRP3 inflammasome activity) were used to pre-treat A549 cells before ZnO-NPs stimulation in the in vitro experiments, respectively. The results from this study suggested that ZnO-NP-induced ROS production triggered the accumulation of damaged mitochondria and assembly of NLRP3 inflammatory complex, leading to maturation and release of IL-1β. Moreover, ZnO-NP-induced NLRP3 inflammasome activation was partly mediated by mitochondrial damage. Taken together, our study suggested that ZnO-NPs induced ALI through oxidative stress-mediated mitochondrial damage and NLRP3 inflammasome activation and provided insight into the mechanisms of ZnO-NPs-induced ALI.
Collapse
Affiliation(s)
- Lai-Bao Zhuo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yu-Mei Liu
- International School of Public Health and One Health, Hainan Medical University, Haikou, China
| | - Yuhan Jiang
- Department of Built Environment, North Carolina A&T State University, Greensboro, NC, 27411, United States
| | - Zhen Yan
- International School of Public Health and One Health, Hainan Medical University, Haikou, China; School of Public Health, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
26
|
D’Evelyn SM, Bein KJ, Laing EA, Nyguen T, Wu CW, Zhang Q, Pinkerton KE. Short-term and repeated exposure to particulate matter sizes from Imperial Valley, California to induce inflammation and asthmatic-like symptoms in mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:909-927. [PMID: 37698070 PMCID: PMC10550522 DOI: 10.1080/15287394.2023.2257232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Imperial Valley, California has become increasingly hot, dry, and polluted over the past decade. Particulate matter (PM) levels are amongst the highest in this State, associated with significantly higher asthma prevalence among children in the region compared to national and state averages. The present study was performed to test the hypothesis that Imperial Valley PM by size and chemical composition might possess allergenic properties following introduction into murine lungs without prior sensitization to a known allergen with size fraction as a determining factor. In acute exposure experiments, BALB/c male mice were administered a single 50-μl oropharyngeal aspiration of nanopure water (H2O; control) or a stock 1 μg/μl PM solution. In sub-acute exposure experiments, male and female mice were treated with a total of six 16.6-μl intranasal instillations of H2O or stock PM solution over the course of 14 days. In all experiments, pulmonary function tests were performed 24 hr after the final instillation followed by necropsies for the collection of biological samples. Inflammatory responses measured via cellularity in histopathological tissue sections as well as significant, marked influxes of eosinophils and lymphocytes were noted in the bronchoalveolar lavage fluid in mice administered PM compared to control. Allergic responses, including airway hyperresponsiveness and significantly increased expression of IL-1ß, were found in male mice exposed to either PM2.5 or ultrafine (PMUF). A combination of all three size fractions of PM from Imperial Valley initiated atopic and asthmatic-like symptoms in the lungs of mice in the absence of additional allergen or preexisting condition.
Collapse
Affiliation(s)
- Savannah M. D’Evelyn
- Center for Health and the Environment, University of California, Davis, CA, US
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA, US
| | - Keith J. Bein
- Center for Health and the Environment, University of California, Davis, CA, US
- Air Quality Research Center, University of California, Davis, CA, US
| | - Emilia A. Laing
- Center for Health and the Environment, University of California, Davis, CA, US
| | - Tran Nyguen
- Department of Environmental Toxicology, University of California, Davis, CA, US
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, CA, US
| | - Qi Zhang
- Department of Environmental Toxicology, University of California, Davis, CA, US
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, CA, US
| |
Collapse
|
27
|
Scotland BL, Shaw JR, Dharmaraj S, Caprio N, Cottingham AL, Joy Martín Lasola J, Sung JJ, Pearson RM. Cell and biomaterial delivery strategies to induce immune tolerance. Adv Drug Deliv Rev 2023; 203:115141. [PMID: 37980950 PMCID: PMC10842132 DOI: 10.1016/j.addr.2023.115141] [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: 09/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The prevalence of immune-mediated disorders, including autoimmune conditions and allergies, is steadily increasing. However, current therapeutic approaches are often non-specific and do not address the underlying pathogenic condition, often resulting in impaired immunity and a state of generalized immunosuppression. The emergence of technologies capable of selectively inhibiting aberrant immune activation in a targeted, antigen (Ag)-specific manner by exploiting the body's intrinsic tolerance pathways, all without inducing adverse side effects, holds significant promise to enhance patient outcomes. In this review, we will describe the body's natural mechanisms of central and peripheral tolerance as well as innovative delivery strategies using cells and biomaterials targeting innate and adaptive immune cells to promote Ag-specific immune tolerance. Additionally, we will discuss the challenges and future opportunities that warrant consideration as we navigate the path toward clinical implementation of tolerogenic strategies to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Brianna L Scotland
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jacob R Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Shruti Dharmaraj
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Nicholas Caprio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jackline Joy Martín Lasola
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junsik J Sung
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States.
| |
Collapse
|
28
|
Sadrolvaezin A, Pezhman A, Zare I, Nasab SZ, Chamani S, Naghizadeh A, Mostafavi E. Systemic allergic contact dermatitis to palladium, platinum, and titanium: mechanisms, clinical manifestations, prevalence, and therapeutic approaches. MedComm (Beijing) 2023; 4:e386. [PMID: 37873514 PMCID: PMC10590457 DOI: 10.1002/mco2.386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 10/25/2023] Open
Abstract
Contact dermatitis (CD) is an inflammatory skin disease of eczema that is elicited by chemicals or metal ions that have toxic effects without eliciting a T-cell response (contact elicitation) or by small reactive chemicals that modify proteins and induce innate and adaptive immune responses (contact allergens). The clinical condition is characterized by localized skin rash, pruritus, redness, swelling, and lesions, which are mainly detected by patch tests and lymphocyte stimulation. Heavy metals such as palladium (Pd), platinum (Pt), and titanium (Ti) are ubiquitous in our environment. These heavy metals have shown CD effects as allergic agents. Immunological responses result from the interaction of cytokines and T cells. Occupational metal CD accounts for most cases of work-related cutaneous disorders. In this systematic review, the allergic effects of heavy metals, including Pd, Pt, and Ti, and the mechanisms, clinical manifestations, prevalence, and therapeutic approaches are discussed in detail. Furthermore, the therapeutic approaches introduced to treat CD, including corticosteroids, topical calcineurin inhibitors, systemic immunosuppressive agents, phototherapy, and antihistamines, can be effective in the treatment of these diseases in the future. Ultimately, the insights identified could lead to improved therapeutic and diagnostic pathways.
Collapse
Affiliation(s)
- Ali Sadrolvaezin
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Arezou Pezhman
- School of MedicineZahedan Azad University of Medical SciencesZahedanIran
| | - Iman Zare
- Research and Development DepartmentSina Medical Biochemistry Technologies Co. Ltd.ShirazIran
| | - Shima Zahed Nasab
- Department of Life Science EngineeringFaculty of New Sciences and TechnologiesUniversity of TehranTehranIran
| | - Sajad Chamani
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ali Naghizadeh
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
29
|
Xu B, Li S, Shi R, Liu H. Multifunctional mesoporous silica nanoparticles for biomedical applications. Signal Transduct Target Ther 2023; 8:435. [PMID: 37996406 PMCID: PMC10667354 DOI: 10.1038/s41392-023-01654-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 11/25/2023] Open
Abstract
Mesoporous silica nanoparticles (MSNs) are recognized as a prime example of nanotechnology applied in the biomedical field, due to their easily tunable structure and composition, diverse surface functionalization properties, and excellent biocompatibility. Over the past two decades, researchers have developed a wide variety of MSNs-based nanoplatforms through careful design and controlled preparation techniques, demonstrating their adaptability to various biomedical application scenarios. With the continuous breakthroughs of MSNs in the fields of biosensing, disease diagnosis and treatment, tissue engineering, etc., MSNs are gradually moving from basic research to clinical trials. In this review, we provide a detailed summary of MSNs in the biomedical field, beginning with a comprehensive overview of their development history. We then discuss the types of MSNs-based nanostructured architectures, as well as the classification of MSNs-based nanocomposites according to the elements existed in various inorganic functional components. Subsequently, we summarize the primary purposes of surface-functionalized modifications of MSNs. In the following, we discuss the biomedical applications of MSNs, and highlight the MSNs-based targeted therapeutic modalities currently developed. Given the importance of clinical translation, we also summarize the progress of MSNs in clinical trials. Finally, we take a perspective on the future direction and remaining challenges of MSNs in the biomedical field.
Collapse
Affiliation(s)
- Bolong Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Shanshan Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Rui Shi
- National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, 100035, Beijing, China.
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China.
| |
Collapse
|
30
|
Kanika, Khan R. Functionalized nanomaterials targeting NLRP3 inflammasome driven immunomodulation: Friend or Foe. NANOSCALE 2023; 15:15906-15928. [PMID: 37750698 DOI: 10.1039/d3nr03857b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The advancement in drug delivery systems in recent times has significantly enhanced therapeutic effects by enabling site-specific targeting through nanocarriers. These nanocarriers serve as invaluable tools for pharmacotherapeutic advancements against various disorders that enhance the effectiveness of encapsulated drugs by reducing their toxicity and increasing the efficacy of less potent drugs, thereby improving the therapeutic index. Inflammasomes, protein complexes located in the activated immune cell cytoplasm, regulate the activation of caspases involved in inflammation. However, aberrant activation of inflammasomes can result in uncontrolled tissue responses, contributing to the development of various diseases. Therefore, achieving a precise balance between inflammasome inhibition and activation is crucial for effectively treating inflammatory disorders through targeted functionalized nanocarriers. Despite the wealth of available data on the relevance of functionalized nanocarriers in inflammatory disorders, the nanotechnological potential to modulate inflammasomes has not been adequately explored. In this comprehensive review, we highlight the latest research on the modulation of the inflammasome cascade, both upregulating and downregulating its function, using nanocarriers in the context of inflammatory disorders. The utilization of nanocarriers as a therapeutic strategy holds immense potential for researchers aiming to effectively target and modulate inflammasomes in the treatment of inflammatory disorders, thus improving disease severity outcomes.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| |
Collapse
|
31
|
Jeon S, Jeon JH, Jeong J, Kim G, Lee S, Kim S, Maruthupandy M, Lee K, Yang SI, Cho WS. Size- and oxidative potential-dependent toxicity of environmentally relevant expanded polystyrene styrofoam microplastics to macrophages. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132295. [PMID: 37597397 DOI: 10.1016/j.jhazmat.2023.132295] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/31/2023] [Accepted: 08/12/2023] [Indexed: 08/21/2023]
Abstract
Expanded polystyrene (EPS), also known as Styrofoam, is a widespread global pollutant, and its lightweight floating property increases its chances of weathering by abrasion and ultraviolet (UV) irradiation, resulting in microplastics. Herein, we investigated the effects of particle size ((1 µm versus 10 µm), UV irradiation (pristine versus UV oxidation), and origin (secondary versus primary) on the toxicity of Styrofoam microplastics. The target cells used in this study were selected based on human exposure-relevant cell lines: differentiated THP-1 cells for macrophages, Caco-2 for enterocytes, HepG2 for hepatocytes, and A549 for alveolar epithelial cells. In the differentiated THP-1 cells, the levels of cytotoxicity and inflammatory cytokines showed size- (1 µm > 10 µm), UV oxidation- (UV > pristine), and origin- (secondary > primary) dependency. Furthermore, the intrinsic oxidative potential of the test particles was positively correlated with cellular oxidative levels and toxicity endpoints, suggesting that the toxicity of Styrofoam microplastics also follows the oxidative stress paradigm. Additionally, all microplastics induced the activation of the pyrin domain-containing protein 3 (NLRP3) inflammasome and the release of interleukin-1β (IL-1β). These results imply that weathering process can aggravate the toxicity of Styrofoam microplastics due to the increased oxidative potential and decreased particle size.
Collapse
Affiliation(s)
- Soyeon Jeon
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Jun Hui Jeon
- Department of Applied Chemistry, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Jiyoung Jeong
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Gyuri Kim
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Sinuk Lee
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Songyeon Kim
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Muthuchamy Maruthupandy
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do 56212, Republic of Korea
| | - Sung Ik Yang
- Department of Applied Chemistry, Kyung Hee University, Yongin-si 17104, Republic of Korea.
| | - Wan-Seob Cho
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea.
| |
Collapse
|
32
|
Gupta G, Kaur J, Bhattacharya K, Chambers BJ, Gazzi A, Furesi G, Rauner M, Fuoco C, Orecchioni M, Delogu LG, Haag L, Stehr JE, Thomen A, Bordes R, Malmberg P, Seisenbaeva GA, Kessler VG, Persson M, Fadeel B. Exploiting Mass Spectrometry to Unlock the Mechanism of Nanoparticle-Induced Inflammasome Activation. ACS NANO 2023; 17:17451-17467. [PMID: 37643371 PMCID: PMC10510732 DOI: 10.1021/acsnano.3c05600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Nanoparticles (NPs) elicit sterile inflammation, but the underlying signaling pathways are poorly understood. Here, we report that human monocytes are particularly vulnerable to amorphous silica NPs, as evidenced by single-cell-based analysis of peripheral blood mononuclear cells using cytometry by time-of-flight (CyToF), while silane modification of the NPs mitigated their toxicity. Using human THP-1 cells as a model, we observed cellular internalization of silica NPs by nanoscale secondary ion mass spectrometry (nanoSIMS) and this was confirmed by transmission electron microscopy. Lipid droplet accumulation was also noted in the exposed cells. Furthermore, time-of-flight secondary ion mass spectrometry (ToF-SIMS) revealed specific changes in plasma membrane lipids, including phosphatidylcholine (PC) in silica NP-exposed cells, and subsequent studies suggested that lysophosphatidylcholine (LPC) acts as a cell autonomous signal for inflammasome activation in the absence of priming with a microbial ligand. Moreover, we found that silica NPs elicited NLRP3 inflammasome activation in monocytes, whereas cell death transpired through a non-apoptotic, lipid peroxidation-dependent mechanism. Together, these data further our understanding of the mechanism of sterile inflammation.
Collapse
Affiliation(s)
- Govind Gupta
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jasreen Kaur
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Kunal Bhattacharya
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Arianna Gazzi
- Department
of Biomedical Sciences, University of Padua, Padua 35121, Italy
| | - Giulia Furesi
- Department
of Medicine III and Center for Healthy Aging, TU Dresden, 01307 Dresden, Germany
| | - Martina Rauner
- Department
of Medicine III and Center for Healthy Aging, TU Dresden, 01307 Dresden, Germany
| | - Claudia Fuoco
- Department
of Biology, University of Rome Tor Vergata, Rome 00173, Italy
| | - Marco Orecchioni
- Division
of Inflammation Biology, La Jolla Institute
for Immunology, La Jolla, California 92037, United States
| | - Lucia Gemma Delogu
- Department
of Biomedical Sciences, University of Padua, Padua 35121, Italy
| | - Lars Haag
- Department
of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Jan Eric Stehr
- Department
of Physics, Chemistry and Biology, Linköping
University, 581 83 Linköping, Sweden
| | - Aurélien Thomen
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 412 96 Göteborg, Sweden
| | - Romain Bordes
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, 412 96 Göteborg, Sweden
| | - Per Malmberg
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, 412 96 Göteborg, Sweden
| | - Gulaim A. Seisenbaeva
- Department
of Molecular Sciences, Swedish University
of Agricultural Sciences, 750 07 Uppsala, Sweden
| | - Vadim G. Kessler
- Department
of Molecular Sciences, Swedish University
of Agricultural Sciences, 750 07 Uppsala, Sweden
| | - Michael Persson
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, 412 96 Göteborg, Sweden
| | - Bengt Fadeel
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
33
|
Pan Y, Ikoma K, Matsui R, Nakayama A, Takemura N, Saitoh T. Dasatinib suppresses particulate-induced pyroptosis and acute lung inflammation. Front Pharmacol 2023; 14:1250383. [PMID: 37705538 PMCID: PMC10495768 DOI: 10.3389/fphar.2023.1250383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Background: Humans are constantly exposed to various industrial, environmental, and endogenous particulates that result in inflammatory diseases. After being engulfed by immune cells, viz. Macrophages, such particulates lead to phagolysosomal dysfunction, eventually inducing pyroptosis, a form of cell death accompanied by the release of inflammatory mediators, including members of the interleukin (IL)-1 family. Phagolysosomal dysfunction results in the activation of the nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, an immune complex that induces pyroptosis upon exposure to various external stimuli. However, several particulates induce pyroptosis even if the NLRP3 inflammasome is inhibited; this indicates that such inhibition is not always effective in treating diseases induced by particulates. Therefore, discovery of drugs suppressing particulate-induced NLRP3-independent pyroptosis is warranted. Methods: We screened compounds that inhibit silica particle (SP)-induced cell death and release of IL-1α using RAW264.7 cells, which are incapable of NLRP3 inflammasome formation. The candidates were tested for their ability to suppress particulate-induced pyroptosis and phagolysosomal dysfunction using mouse primary macrophages and alleviate SP-induced NLRP3-independent lung inflammation. Results: Several Src family kinase inhibitors, including dasatinib, effectively suppressed SP-induced cell death and IL-1α release. Furthermore, dasatinib suppressed pyroptosis induced by other particulates but did not suppress that induced by non-particulates, such as adenosine triphosphate. Dasatinib reduced SP-induced phagolysosomal dysfunction without affecting phagocytosis of SPs. Moreover, dasatinib treatment strongly suppressed the increase in IL-1α levels and neutrophil counts in the lungs after intratracheal SP administration. Conclusion: Dasatinib suppresses particulate-induced pyroptosis and can be used to treat relevant inflammatory diseases.
Collapse
Affiliation(s)
- Yixi Pan
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kenta Ikoma
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Risa Matsui
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Naoki Takemura
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka, Japan
| |
Collapse
|
34
|
Pavan C, Santalucia R, Escolano-Casado G, Ugliengo P, Mino L, Turci F. Physico-Chemical Approaches to Investigate Surface Hydroxyls as Determinants of Molecular Initiating Events in Oxide Particle Toxicity. Int J Mol Sci 2023; 24:11482. [PMID: 37511241 PMCID: PMC10380507 DOI: 10.3390/ijms241411482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The study of molecular recognition patterns is crucial for understanding the interactions between inorganic (nano)particles and biomolecules. In this review we focus on hydroxyls (OH) exposed at the surface of oxide particles (OxPs) which can play a key role in molecular initiating events leading to OxPs toxicity. We discuss here the main analytical methods available to characterize surface OH from a quantitative and qualitative point of view, covering thermogravimetry, titration, ζ potential measurements, and spectroscopic approaches (NMR, XPS). The importance of modelling techniques (MD, DFT) for an atomistic description of the interactions between membranes/proteins and OxPs surfaces is also discussed. From this background, we distilled a new approach methodology (NAM) based on the combination of IR spectroscopy and bioanalytical assays to investigate the molecular interactions of OxPs with biomolecules and membranes. This NAM has been already successfully applied to SiO2 particles to identify the OH patterns responsible for the OxPs' toxicity and can be conceivably extended to other surface-hydroxylated oxides.
Collapse
Affiliation(s)
- Cristina Pavan
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Torino, 10125 Torino, Italy
- Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Rosangela Santalucia
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10125 Torino, Italy
| | - Guillermo Escolano-Casado
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10125 Torino, Italy
| | - Piero Ugliengo
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10125 Torino, Italy
| | - Lorenzo Mino
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10125 Torino, Italy
| | - Francesco Turci
- Department of Chemistry, University of Torino, Via Giuria 7, 10125 Torino, Italy
- "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Torino, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10125 Torino, Italy
| |
Collapse
|
35
|
Zhao Y, Liu S, Xu H. Effects of microplastic and engineered nanomaterials on inflammatory bowel disease: A review. CHEMOSPHERE 2023; 326:138486. [PMID: 36963581 DOI: 10.1016/j.chemosphere.2023.138486] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 06/18/2023]
Abstract
Many microplastics and engineered nanomaterials (ENMs) exist in the daily environment. The intestinal impact of these exogenous fine particles on inflammatory bowel disease (IBD) people may be unpredictable. In this paper, we reviewed the recent progress in the effect of microplastics and ENMs on IBD individuals. We also compared and summarized the various roles of microplastics and ENMs in healthy and IBD bodies, including factors such as particle size, particle properties, intestinal microenvironment, interaction with the intestinal barrier, and molecular mechanism. Our literature review showed that microplastics could be accomplices in the development of IBD and could cause severe intestinal inflammation. Moreover, ENMs could elicit diverse exposure outcomes in healthy and IBD bodies. Silicon dioxide nanoparticles (SiO2 NPs), titanium dioxide nanoparticles (TiO2 NPs), and graphene oxide (GO) displayed slight to adverse effects that turned into apparent adverse effects, while zinc oxide nanoparticles (ZnO NPs) and silver nanoparticles (Ag NPs) showed a toxic effect that became therapeutic. A deeper understanding of the impact of microplastics and ENMs on the high-risk group was needed, and we proposed several insights into the research priorities and directions.
Collapse
Affiliation(s)
- Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
36
|
Singh R, Srivastava P, Manna PP. Chemokine-targeted nanoparticles: stimulation of the immune system in cancer immunotherapy. EXPLORATION OF IMMUNOLOGY 2023:123-147. [DOI: 10.37349/ei.2023.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/24/2023] [Indexed: 01/06/2025]
Abstract
Surgery, chemotherapy, radiation therapy, and immunotherapy are potential therapeutic choices for many malignant and metastatic cancers. Despite adverse side effects and pain, surgery and chemotherapy continue to be the most common cancer treatments. However, patients treated with immunotherapy had better cancer control than those who got other treatments. There are two methods to activate immunological pathways: systemically and locally. To modify the tumor microenvironment (TME), the former uses systemic cytokine/chemokine (CK) delivery, whilst the latter uses immunological checkpoints or small molecule inhibitors. Organic and inorganic nanomaterials (NMs) enhanced the efficacy of cancer immunotherapy. NMs can transmit drugs, peptides, antigens, antibodies, whole cell membranes, etc. Surface-modified NMs precisely target and enter the tissues. The inner core of surface-modified NMs is composed of chemicals with limited bioavailability and biocompatibility, resulting in prolonged blood retention and decreased renal clearance. These platforms hinder or prevent many immune cell activities and modify the TME, enhancing the efficiency of cancer immunotherapy. By inhibiting CK/CK receptor signaling, cell migration and other immune responses could be controlled. Developing CK-targeted nanoparticles (NPs) that inhibit CK signaling or take advantage of the ligand-receptor connection is possible. Surface chemical modification of NMs with CKs or specific peptides has several medicinal applications, including tissue-specific drug delivery and limited cell migration in cancer-afflicted conditions. This review covers current developments in the role of different groups of CK-loaded NP in tumor therapy targeting immune cells and cancer. It also covers the role of NP targeting CK signaling which aids in immunogenic cell death (ICD) and induction of antitumor immunity. In addition, CK gene silencing and its capacity to prevent cancer metastasis as well as inhibition of immune cell migration to modulate the TME are discussed.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Prateek Srivastava
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India;Current address: Postdoctoral Fellow, Ben-Gurion University of the Negev, Beersheba 8410501, Israel
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
37
|
Alijagic A, Hedbrant A, Persson A, Larsson M, Engwall M, Särndahl E. NLRP3 inflammasome as a sensor of micro- and nanoplastics immunotoxicity. Front Immunol 2023; 14:1178434. [PMID: 37143682 PMCID: PMC10151538 DOI: 10.3389/fimmu.2023.1178434] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Micro- and nanoplastics (MNPs) are emerging pollutants with scarcely investigated effects on human innate immunity. If they follow a similar course of action as other, more thoroughly investigated particulates, MNPs may penetrate epithelial barriers, potentially triggering a cascade of signaling events leading to cell damage and inflammation. Inflammasomes are intracellular multiprotein complexes and stimulus-induced sensors critical for mounting inflammatory responses upon recognition of pathogen- or damage-associated molecular patterns. Among these, the NLRP3 inflammasome is the most studied in terms of activation via particulates. However, studies delineating the ability of MNPs to affect NLRP3 inflammasome activation are still rare. In this review, we address the issue of MNPs source and fate, highlight the main concepts of inflammasome activation via particulates, and explore recent advances in using inflammasome activation for assessment of MNP immunotoxicity. We also discuss the impact of co-exposure and MNP complex chemistry in potential inflammasome activation. Development of robust biological sensors is crucial in order to maximize global efforts to effectively address and mitigate risks that MNPs pose for human health.
Collapse
Affiliation(s)
- Andi Alijagic
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Alexander Persson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
38
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
39
|
Coutinho Almeida-da-Silva CL, Cabido LF, Chin WC, Wang G, Ojcius DM, Li C. Interactions between silica and titanium nanoparticles and oral and gastrointestinal epithelia: Consequences for inflammatory diseases and cancer. Heliyon 2023; 9:e14022. [PMID: 36938417 PMCID: PMC10020104 DOI: 10.1016/j.heliyon.2023.e14022] [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: 10/27/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/21/2023] Open
Abstract
Engineered nanoparticles (NPs) composed of elements such as silica and titanium, smaller than 100 nm in diameter and their aggregates, are found in consumer products such as cosmetics, food, antimicrobials and drug delivery systems, and oral health products such as toothpaste and dental materials. They may also interact accidently with epithelial tissues in the intestines and oral cavity, where they can aggregate into larger particles and induce inflammation through pathways such as inflammasome activation. Persistent inflammation can lead to precancerous lesions. Both the particles and lesions are difficult to detect in biopsies, especially in clinical settings that screen large numbers of patients. As diagnosis of early stages of disease can be lifesaving, there is growing interest in better understanding interactions between NPs and epithelium and developing rapid imaging techniques that could detect foreign particles and markers of inflammation in epithelial tissues. NPs can be labelled with fluorescence or radioactive isotopes, but it is challenging to detect unlabeled NPs with conventional imaging techniques. Different current imaging techniques such as synchrotron radiation X-ray fluorescence spectroscopy are discussed here. Improvements in imaging techniques, coupled with the use of machine learning tools, are needed before diagnosis of particles in biopsies by automated imaging could move usefully into the clinic.
Collapse
Affiliation(s)
| | - Leticia Ferreira Cabido
- Department of Oral and Maxillofacial Surgery, University of the Pacific, San Francisco, CA, USA
| | - Wei-Chun Chin
- Department of Bioengineering, University of California, Merced, CA, USA
| | - Ge Wang
- Department of Biomedical Engineering, Biomedical Imaging Center, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, San Francisco, CA, USA
| | - Changqing Li
- Department of Bioengineering, University of California, Merced, CA, USA
| |
Collapse
|
40
|
Borgmann LM, Johnsen S, Santos de Oliveira C, Martins de Souza E Silva J, Li J, Kirchlechner C, Gomard G, Wiegand G, Hölscher H. Porous polymeric microparticles foamed with supercritical CO 2as scattering white pigments. BIOINSPIRATION & BIOMIMETICS 2023; 18:026011. [PMID: 36731134 DOI: 10.1088/1748-3190/acb899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Nowadays, titanium dioxide (TiO2) is the most commercially relevant white pigment. Nonetheless, it is widely criticized due to its energy-intensive extraction and costly disposal of harmful by-products. Furthermore, recent studies discuss its potential harm for the environment and the human health. Environment-friendly strategies for the replacement of TiO2as a white pigment can be inspired from nature. Here whiteness often originates from broadband light scattering air cavities embedded in materials with refractive indices much lower than that of TiO2. Such natural prototypes can be mimicked by introducing air-filled nano-scale cavities into commonly used polymers. Here, we demonstrate the foaming of initially transparent poly(methyl methacrylate) (PMMA) microspheres with non-toxic, inert, supercritical CO2. The properties of the foamed, white polymeric pigments with light scattering nano-pores are evaluated as possible replacement for TiO2pigments. For that, the inner foam structure of the particles was imaged by phase-contrast x-ray nano-computed tomography (nano-CT), the optical properties were evaluated via spectroscopic measurements, and the mechanical stability was examined by micro compression experiments. Adding a diffusion barrier surrounding the PMMA particles during foaming allows to extend the foaming process towards smaller particles. Finally, we present a basic white paint prototype as exemplary application.
Collapse
Affiliation(s)
- Luisa Maren Borgmann
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Siegbert Johnsen
- Institute of Catalysis Research and Technology (IKFT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | | | - Juliana Martins de Souza E Silva
- Martin-Luther-Universität Halle-Wittenberg, Institute of Physics, 06120 Halle (Saale), Germany
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, 06120 Halle (Saale), Germany
| | - Juan Li
- Institute for Applied Materials (IAM), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Christoph Kirchlechner
- Institute for Applied Materials (IAM), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Guillaume Gomard
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
- Carl Zeiss AG, Zeiss Innovation Hub, Hermann-von-Helmholtz-Platz 6, 76344 Eggenstein-Leopoldshafen, Germany
| | - Gabriele Wiegand
- Institute of Catalysis Research and Technology (IKFT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Hendrik Hölscher
- Institute of Microstructure Technology (IMT), Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| |
Collapse
|
41
|
Huang W, Zhang Z, Qiu Y, Gao Y, Fan Y, Wang Q, Zhou Q. NLRP3 inflammasome activation in response to metals. Front Immunol 2023; 14:1055788. [PMID: 36845085 PMCID: PMC9950627 DOI: 10.3389/fimmu.2023.1055788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Implant surgery is followed by a series of inflammatory reactions that directly affect its postoperative results. The inflammasome plays a vital role in the inflammatory response by inducing pyroptosis and producing interleukin-1β, which plays a critical role in inflammation and tissue damage. Therefore, it is essential to study the activation of the inflammasome in the bone healing process after implant surgery. As metals are the primary implant materials, metal-induced local inflammatory reactions have received significant attention, and there has been more and more research on the activation of the NLRP3 (NOD-like receptor protein-3) inflammasome caused by these metals. In this review, we consolidate the basic knowledge on the NLRP3 inflammasome structures, the present knowledge on the mechanisms of NLRP3 inflammasome activation, and the studies of metal-induced NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Wanyi Huang
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Ziqi Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Yueyang Qiu
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Yuan Gao
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
- Department of Orthodontics, Shenyang Stomatological Hospital, Shenyang, China
| | - Yongqiang Fan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qing Zhou
- School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| |
Collapse
|
42
|
Boraschi D, Canesi L, Drobne D, Kemmerling B, Pinsino A, Prochazkova P. Interaction between nanomaterials and the innate immune system across evolution. Biol Rev Camb Philos Soc 2023; 98:747-774. [PMID: 36639936 DOI: 10.1111/brv.12928] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 01/15/2023]
Abstract
Interaction of engineered nanomaterials (ENMs) with the immune system mainly occurs with cells and molecules of innate immunity, which are present in interface tissues of living organisms. Immuno-nanotoxicological studies aim at understanding if and when such interaction is inconsequential or may cause irreparable damage. Since innate immunity is the first line of immune reactivity towards exogenous agents and is highly conserved throughout evolution, this review focuses on the major effector cells of innate immunity, the phagocytes, and their major sensing receptors, Toll-like receptors (TLRs), for assessing the modes of successful versus pathological interaction between ENMs and host defences. By comparing the phagocyte- and TLR-dependent responses to ENMs in plants, molluscs, annelids, crustaceans, echinoderms and mammals, we aim to highlight common recognition and elimination mechanisms and the general sufficiency of innate immunity for maintaining tissue integrity and homeostasis.
Collapse
Affiliation(s)
- Diana Boraschi
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Science (CAS), 1068 Xueyuan Blvd, 518071, Shenzhen, China.,Institute of Protein Biochemistry and Cell Biology (IBBC), CNR, Via Pietro Castellino 111, 80131, Naples, Italy.,Stazione Zoologica Anton Dohrn (SZN), Villa Comunale, 80132, Napoli, Italy.,China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (SIAT, CNR, SZN), Napoli, Italy
| | - Laura Canesi
- Department of Earth, Environment and Life Sciences, University of Genova, Corso Europa 26, 16132, Genova, Italy
| | - Damjana Drobne
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000, Ljubliana, Slovenia
| | - Birgit Kemmerling
- ZMBP - Center for Plant Molecular Biology, Plant Biochemistry, University of Tübingen, Auf der Morgenstelle 32, 72076, Tübingen, Germany
| | - Annalisa Pinsino
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Ugo La Malfa 153, 90146, Palermo, Italy
| | - Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| |
Collapse
|
43
|
Takemura N. [Development of Anti-inflammatory Drugs with Novel Mechanisms of Action Targeting Pyroptosis]. YAKUGAKU ZASSHI 2023; 143:997-1003. [PMID: 38044115 DOI: 10.1248/yakushi.23-00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Programmed cell death plays various physiological roles, one of which is an immune response that protects the body from infectious pathogens such as bacteria and viruses. Pathogen infection causes dysfunction of cellular organelles, such as mitochondria and lysosomes, triggering stress signals that induce programmed cell death. In some cases, cell death coincides with intracellular inflammatory cytokine release. Such programmed cell death, accompanied by the induction of inflammatory responses, is called pyroptosis, which inhibits pathogen proliferation within cells and attracts leukocytes that eliminate the pathogens, thereby preventing infection spread. Additionally, pyroptosis can be induced by noninfectious stimuli such as drugs, pollutants, and nutrients, resulting in severe inflammatory disease. Therefore, the development of effective anti-inflammatory drugs that prevent pyroptosis based on the understanding of the mechanisms responsible for its induction is an urgent requirement. This review provides an overview of the non-infectious inflammatory response caused by pyroptosis and the development of new anti-inflammatory drugs that target organelles to prevent pyroptosis to treat relevant inflammatory diseases.
Collapse
Affiliation(s)
- Naoki Takemura
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
44
|
Yang WK, Kim SW, Youn SH, Hyun SH, Han CK, Park YC, Lee YC, Kim SH. Respiratory protective effects of Korean Red Ginseng in a mouse model of particulate matter 4-induced airway inflammation. J Ginseng Res 2023; 47:81-88. [PMID: 36644393 PMCID: PMC9834024 DOI: 10.1016/j.jgr.2022.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/25/2022] [Accepted: 05/23/2022] [Indexed: 01/18/2023] Open
Abstract
Background Air pollution has led to an increased exposure of all living organisms to fine dust. Therefore, research efforts are being made to devise preventive and therapeutic remedies against fine dust-induced chronic diseases. Methods Research of the respiratory protective effects of KRG extract in a particulate matter (PM; aerodynamic diameter of <4 μm) plus diesel exhaust particle (DEP) (PM4+D)-induced airway inflammation model. Nitric oxide production, expression of pro-inflammatory mediators and cytokines, and IRAK-1, TAK-1, and MAPK pathways were examined in PM4-stimulated MH-S cells. BALB/c mice exposed to PM4+D mixture by intranasal tracheal injection three times a day for 12 days at 3 day intervals and KRGE were administered orally for 12 days. Histological of lung and trachea, and immune cell subtype analyses were performed. Expression of pro-inflammatory mediators and cytokines in bronchoalveolar lavage fluid (BALF) and lung were measured. Immunohistofluorescence staining for IRAK-1 localization in lung were also evaluated. Results KRGE inhibited the production of nitric oxide, the expression of pro-inflammatory mediators and cytokines, and expression and phosphorylation of all downstream factors of NF-κB, including IRAK-1 and MAPK/AP1 pathway in PM4-stimulated MH-S cells. KRGE suppressed inflammatory cell infiltration and number of immune cells, histopathologic damage, and inflammatory symptoms in the BALF and lungs induced by PM4+D; these included increased alveolar wall thickness, accumulation of collagen fibers, and TNF-α, MIP2, CXCL-1, IL-1α, and IL-17 cytokine release. Moreover, PM4 participates induce alveolar macrophage death and interleukin-1α release by associating with IRAK-1 localization was also potently inhibited by KRGE in the lungs of PM4+D-induced airway inflammation model. KRGE suppresses airway inflammatory responses, including granulocyte infiltration into the airway, by regulating the expression of chemokines and inflammatory cytokines via inhibition of IRAK-1 and MAPK pathway. Conclusion: Our results indicate the potential of KRGE to serve as an effective therapeutic agent against airway inflammation and respiratory diseases.
Collapse
Affiliation(s)
- Won-Kyung Yang
- Division of Respiratory Medicine, Department of Internal Medicine, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, Republic of Korea
| | - Sung-Won Kim
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Soo Hyun Youn
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Sun Hee Hyun
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Chang-Kyun Han
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Yang-Chun Park
- Division of Respiratory Medicine, Department of Internal Medicine, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Young-Cheol Lee
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju, Republic of Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, Republic of Korea
- Corresponding author. Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, 34520, Republic of Korea.
| |
Collapse
|
45
|
Reichinger D, Reithofer M, Hohagen M, Drinic M, Tobias J, Wiedermann U, Kleitz F, Jahn-Schmid B, Becker CFW. A Biomimetic, Silaffin R5-Based Antigen Delivery Platform. Pharmaceutics 2022; 15:pharmaceutics15010121. [PMID: 36678751 PMCID: PMC9866965 DOI: 10.3390/pharmaceutics15010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023] Open
Abstract
Nature offers a wide range of evolutionary optimized materials that combine unique properties with intrinsic biocompatibility and that can be exploited as biomimetic materials. The R5 and RRIL peptides employed here are derived from silaffin proteins that play a crucial role in the biomineralization of marine diatom silica shells and are also able to form silica materials in vitro. Here, we demonstrate the application of biomimetic silica particles as a vaccine delivery and adjuvant platform by linking the precipitating peptides R5 and the RRIL motif to a variety of peptide antigens. The resulting antigen-loaded silica particles combine the advantages of biomaterial-based vaccines with the proven intracellular uptake of silica particles. These particles induce NETosis in human neutrophils as well as IL-6 and TNF-α secretion in murine bone marrow-derived dendritic cells.
Collapse
Affiliation(s)
- Daniela Reichinger
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Manuel Reithofer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180 Vienna, Austria
| | - Mariam Hohagen
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
- Department of Inorganic Chemistry–Functional Materials, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Mirjana Drinic
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
| | - Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
| | - Freddy Kleitz
- Department of Inorganic Chemistry–Functional Materials, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Beatrice Jahn-Schmid
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Christian F. W. Becker
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 38, 1090 Vienna, Austria
- Correspondence:
| |
Collapse
|
46
|
Danso IK, Woo JH, Lee K. Pulmonary Toxicity of Polystyrene, Polypropylene, and Polyvinyl Chloride Microplastics in Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227926. [PMID: 36432032 PMCID: PMC9694469 DOI: 10.3390/molecules27227926] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Globally, plastics are used in various products. Concerns regarding the human body's exposure to plastics and environmental pollution have increased with increased plastic use. Microplastics can be detected in the atmosphere, leading to potential human health risks through inhalation; however, the toxic effects of microplastic inhalation are poorly understood. In this study, we examined the pulmonary toxicity of polystyrene (PS), polypropylene (PP), and polyvinyl chloride (PVC) in C57BL/6, BALB/c, and ICR mice strains. Mice were intratracheally instilled with 5 mg/kg of PS, PP, or PVC daily for two weeks. PS stimulation increased inflammatory cells in the bronchoalveolar lavage fluid (BALF) of C57BL/6 and ICR mice. Histopathological analysis of PS-instilled C57BL/6 and PP-instilled ICR mice showed inflammatory cell infiltration. PS increased the NLR family pyrin domain containing 3 (NLRP3) inflammasome components in the lung tissue of C57BL/6 and ICR mice, while PS-instilled BALB/c mice remained unchanged. PS stimulation increased inflammatory cytokines, including IL-1β and IL-6, in BALF of C57BL/6 mice. PP-instilled ICR mice showed increased NLRP3, ASC, and Caspase-1 in the lung tissue compared to the control groups and increased IL-1β levels in BALF. These results could provide baseline data for understanding the pulmonary toxicity of microplastic inhalation.
Collapse
Affiliation(s)
- Isaac Kwabena Danso
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baekhak 1-gil, Jeongeup 56212, Jeollabuk-do, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Jong-Hwan Woo
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baekhak 1-gil, Jeongeup 56212, Jeollabuk-do, Republic of Korea
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baekhak 1-gil, Jeongeup 56212, Jeollabuk-do, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea
- Correspondence: ; Tel.: +82-63-570-8740
| |
Collapse
|
47
|
Zhang G, Luo W, Yang W, Li S, Li D, Zeng Y, Li Y. The importance of the
IL
‐1 family of cytokines in nanoimmunosafety and nanotoxicology. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1850. [DOI: 10.1002/wnan.1850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Guofang Zhang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenhe Luo
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenjie Yang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Su Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Dongjie Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yanqiao Zeng
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
48
|
Waheed S, Li Z, Zhang F, Chiarini A, Armato U, Wu J. Engineering nano-drug biointerface to overcome biological barriers toward precision drug delivery. J Nanobiotechnology 2022; 20:395. [PMID: 36045386 PMCID: PMC9428887 DOI: 10.1186/s12951-022-01605-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
The rapid advancement of nanomedicine and nanoparticle (NP) materials presents novel solutions potentially capable of revolutionizing health care by improving efficacy, bioavailability, drug targeting, and safety. NPs are intriguing when considering medical applications because of their essential and unique qualities, including a significantly higher surface to mass ratio, quantum properties, and the potential to adsorb and transport drugs and other compounds. However, NPs must overcome or navigate several biological barriers of the human body to successfully deliver drugs at precise locations. Engineering the drug carrier biointerface can help overcome the main biological barriers and optimize the drug delivery in a more personalized manner. This review discusses the significant heterogeneous biological delivery barriers and how biointerface engineering can promote drug carriers to prevail over hurdles and navigate in a more personalized manner, thus ushering in the era of Precision Medicine. We also summarize the nanomedicines' current advantages and disadvantages in drug administration, from natural/synthetic sources to clinical applications. Additionally, we explore the innovative NP designs used in both non-personalized and customized applications as well as how they can attain a precise therapeutic strategy.
Collapse
Affiliation(s)
- Saquib Waheed
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Zhibin Li
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Fangyingnan Zhang
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Anna Chiarini
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy
| | - Ubaldo Armato
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy
| | - Jun Wu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy.
| |
Collapse
|
49
|
Forster Iii J, Nandi D, Kulkarni A. mRNA-carrying lipid nanoparticles that induce lysosomal rupture activate NLRP3 inflammasome and reduce mRNA transfection efficiency. Biomater Sci 2022; 10:5566-5582. [PMID: 35971974 DOI: 10.1039/d2bm00883a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the last several years, countless developments have been made to engineer more efficient and potent mRNA lipid nanoparticle vaccines, culminating in the rapid development of effective mRNA vaccines against COVID-19. However, despite these advancements and materials approaches, there is still a lack of understanding of the resultant immunogenicity of mRNA lipid nanoparticles. Therefore, a more mechanistic, design-driven approach needs to be taken to determine which biophysical characteristics, especially related to changes in lipid compositions, drive nanoparticle immunogenicity. Here, we synthesized a panel of six mRNA lipid nanoparticle formulations, varying the concentrations of different lipid components and systematically studied their effect on NLRP3 inflammasome activation; a key intracellular protein complex that controls various inflammatory responses. Initial experiments aimed to determine differences in nanoparticle activation of NLRP3 inflammasomes by IL-1β ELISA, which unveiled that nanoparticles with high concentrations of ionizable lipid DLin-MC3-DMA in tandem with high cationic lipid DPTAP and low cholesterol concentration induced the greatest activation of the NLRP3 inflammasome. These results were further corroborated by the measurement of ASC specks indicative of NLRP3 complex assembly, as well as cleaved gasdermin-D and caspase-1 expression indicating complex activation. We also uncovered these activation profiles to be mechanistically correlated primarily with lysosomal rupturing caused by the delayed membrane disruption capabilities of ionizable lipids until the lysosomal stage, as well as by mitochondrial reactive oxygen species (ROS) production and calcium influx for some of the particles. Therefore, we report that the specific, combined effects of each lipid type, most notably ionizable, cationic lipids, and cholesterol, is a crucial mRNA lipid nanoparticle characteristic that varies the endo/lysosomal rupture capabilities of the formulation and activate NLRP3 inflammasomes in a lysosomal rupture dependent manner. These results provide a more concrete understanding of mRNA lipid Nanoparticle-Associated Molecular Patterns for the activation of molecular-level immune responses and provide new lipid composition design considerations for future mRNA-delivery approaches.
Collapse
Affiliation(s)
- James Forster Iii
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA. .,Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA. .,Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA.,Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
50
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|