Figure - available from: Nature Nanotechnology
This content is subject to copyright. Terms and conditions apply.
Siglec-14-Syk-mediated inflammatory responses to MWCNTs are blocked by fostamatinib
a, Syk null cells were generated by CRISPR/Cas9-mediated targeting and were cloned by limiting dilution. Syk expression was analysed by immunoblot. b, Sequence of Syk in WT cells and mutant clone #3 alleles around the target locus. The gRNA target sequence is in bold. Deleted bases are indicated by hyphens. c, Cell surface expression of Siglec-14 on the indicated THP-1 cells was analysed as in Fig. 3b. d, Phagocytosis of MWCNTs by the indicated THP-1 cells was analysed as in Fig. 3e. e, IL-1β secretion from the indicated THP-1 cells was analysed as in Fig. 3g. Data are shown as mean ± s.d. (n = 3). ***P < 0.001, one-way ANOVA with Tukey–Kramer test. f, PMA-primed Siglec-14/THP-1 cells were pretreated with the indicated dose of R406 for 1 h and then were treated with MWCNTs (30 μg ml⁻¹) or nigericin (3 μM) for 5 h. The percentage reduction of IL-1β secretion was calculated as the amount of IL-1β produced by the indicated dose of R406-treated cells × 100/the amount of IL-1β produced by R406-untreated cells. Data are shown as mean ± s.d. (n = 3). **P = 0.0042, ***P < 0.001, one-way ANOVA with Tukey–Kramer test. g, LPS-primed S14+/− donor PBMCs (n = 4) were pretreated the indicated dose of R406 for 1 h and then were treated with MWCNTs (10 μg ml⁻¹) or ATP (1 mM) for 3 h. The percentage reduction of IL-1β secretion in individuals was calculated as in f. Data are shown as mean ± s.d. (n = 4). ***P < 0.01, one-way ANOVA with Tukey–Kramer test. h, Mock- or Siglec-14-transduced mice (n = 6 each) generated as in Fig. 3h were orally administered with R788 (0.6 mg per head) or vehicle (0.5% w/v methyl cellulose 400 solution) at 12 h and 0.5 h before intratracheal injection of MWCNTs (50 μg per head). One day later, the concentration of IL-1β and TNF-α in BALF was measured by ELISA. *P = 0.0267, ***P < 0.001, two-way ANOVA with Tukey–Kramer test.
Source data

Siglec-14-Syk-mediated inflammatory responses to MWCNTs are blocked by fostamatinib a, Syk null cells were generated by CRISPR/Cas9-mediated targeting and were cloned by limiting dilution. Syk expression was analysed by immunoblot. b, Sequence of Syk in WT cells and mutant clone #3 alleles around the target locus. The gRNA target sequence is in bold. Deleted bases are indicated by hyphens. c, Cell surface expression of Siglec-14 on the indicated THP-1 cells was analysed as in Fig. 3b. d, Phagocytosis of MWCNTs by the indicated THP-1 cells was analysed as in Fig. 3e. e, IL-1β secretion from the indicated THP-1 cells was analysed as in Fig. 3g. Data are shown as mean ± s.d. (n = 3). ***P < 0.001, one-way ANOVA with Tukey–Kramer test. f, PMA-primed Siglec-14/THP-1 cells were pretreated with the indicated dose of R406 for 1 h and then were treated with MWCNTs (30 μg ml⁻¹) or nigericin (3 μM) for 5 h. The percentage reduction of IL-1β secretion was calculated as the amount of IL-1β produced by the indicated dose of R406-treated cells × 100/the amount of IL-1β produced by R406-untreated cells. Data are shown as mean ± s.d. (n = 3). **P = 0.0042, ***P < 0.001, one-way ANOVA with Tukey–Kramer test. g, LPS-primed S14+/− donor PBMCs (n = 4) were pretreated the indicated dose of R406 for 1 h and then were treated with MWCNTs (10 μg ml⁻¹) or ATP (1 mM) for 3 h. The percentage reduction of IL-1β secretion in individuals was calculated as in f. Data are shown as mean ± s.d. (n = 4). ***P < 0.01, one-way ANOVA with Tukey–Kramer test. h, Mock- or Siglec-14-transduced mice (n = 6 each) generated as in Fig. 3h were orally administered with R788 (0.6 mg per head) or vehicle (0.5% w/v methyl cellulose 400 solution) at 12 h and 0.5 h before intratracheal injection of MWCNTs (50 μg per head). One day later, the concentration of IL-1β and TNF-α in BALF was measured by ELISA. *P = 0.0267, ***P < 0.001, two-way ANOVA with Tukey–Kramer test. Source data

Source publication
Article
Full-text available
For the design and development of innovative carbon nanotube (CNT)-based tools and applications, an understanding of the molecular interactions between CNTs and biological systems is essential. In this study, a three-dimensional protein-structure-based in silico screen identified the paired immune receptors, sialic acid immunoglobulin-like binding...

Citations

... cNt exposure can cause oxidative stress via overproduction of ROS in cells, leading to cytotoxicity [81,[91][92][93]. they can activate inflammatory pathways, triggering the release of pro-inflammatory cytokines and chemokines [94][95][96][97]. they can directly damage DNA or indirectly induce DNA damage (genotoxicity) through ROS production, potentially leading to mutations and cancer development [98,99]. ...
Article
Carbon nanotubes (CNTs) are allotropes of carbon, composed of carbon atoms forming a tube-like structure. Their high surface area, chemical stability, and rich electronic polyaromatic structure facilitate their drug-carrying capacity. Therefore, CNTs have been intensively explored for several biomedical applications, including as a potential treatment option for cancer. By incorporating smart fabrication strategies, CNTs can be designed to specifically target cancer cells. This targeted drug delivery approach not only maximizes the therapeutic utility of CNTs but also minimizes any potential side effects of free drug molecules. CNTs can also be utilized for photothermal therapy (PTT) which uses photosensitizers to generate reactive oxygen species (ROS) to kill cancer cells, and in immunotherapeutic applications. Regarding the latter, for example, CNT-based formulations can preferentially target intra-tumoural regulatory t-cells. CNTs also act as efficient antigen presenters. with their capabilities for photoacoustic, fluorescent and Raman imaging, CNTs are excellent diagnostic tools as well. Further, metallic nanoparticles, such as gold or silver nanoparticles, are combined with CNTs to create nanobiosensors to measure biological reactions. This review focuses on current knowledge about the theranostic potential of CNTs, challenges associated with their large-scale production, their possible side effects and important parameters to consider when exploring their clinical usage.
... Cytotoxicity: cell death PARP (apoptosis) 182 , RIP (necrosis) 182 , GSDMD (pyroptosis) 183 , caspase 183 Autophagy activation 184 Genotoxicity γH2AX detection 185,186 Micronucleus assay 187,188 Immune response Pro-inflammatory cytokine measurement, such as IL-1β, IL-4, TNF 117,189,190 Activation of pattern recognition receptor pathways, such as NLRP3 inflammasome activation 117,191 Inflammatory markers, such as white blood cell count, neutrophils, lymphocytes, monocytes, eosinophils 67 Immune cell activation 190 RNS and ROS measurement 116,180,186 Oxidative stress Acellular oxidative potential assay 192 RNS and ROS measurement 116,180,186 Cell/organelle morphology Bright-field microscopy TEM 186,193 Membrane damage by l-leucyl-l-leucin methyl ester 189,194 Absorption, distribution, metabolism and excretion NIR fluorescence imaging 11 Raman mapping 195 Confocal fluorescence microscopy 196,197 In vivo toxicity Survival 67 Weight loss 67 Tissue-or organ-specific damage, such as serum biomarkers for hepatic injury or renal function 67 Blood oxygenation 67 CNT degradation in biological systems Photoacoustic imaging 162 Differential interference contrast microscopy 162 Dark-field imaging 162,198 Raman spectroscopy 161 toxicological risk. Minimum reporting standards for physicochemical parameters of nanomaterials have been proposed so that occupational safety and health regulators properly assess any toxicological concerns and overcome regulatory hurdles 102,125 . ...
... Cytotoxicity: cell death PARP (apoptosis) 182 , RIP (necrosis) 182 , GSDMD (pyroptosis) 183 , caspase 183 Autophagy activation 184 Genotoxicity γH2AX detection 185,186 Micronucleus assay 187,188 Immune response Pro-inflammatory cytokine measurement, such as IL-1β, IL-4, TNF 117,189,190 Activation of pattern recognition receptor pathways, such as NLRP3 inflammasome activation 117,191 Inflammatory markers, such as white blood cell count, neutrophils, lymphocytes, monocytes, eosinophils 67 Immune cell activation 190 RNS and ROS measurement 116,180,186 Oxidative stress Acellular oxidative potential assay 192 RNS and ROS measurement 116,180,186 Cell/organelle morphology Bright-field microscopy TEM 186,193 Membrane damage by l-leucyl-l-leucin methyl ester 189,194 Absorption, distribution, metabolism and excretion NIR fluorescence imaging 11 Raman mapping 195 Confocal fluorescence microscopy 196,197 In vivo toxicity Survival 67 Weight loss 67 Tissue-or organ-specific damage, such as serum biomarkers for hepatic injury or renal function 67 Blood oxygenation 67 CNT degradation in biological systems Photoacoustic imaging 162 Differential interference contrast microscopy 162 Dark-field imaging 162,198 Raman spectroscopy 161 toxicological risk. Minimum reporting standards for physicochemical parameters of nanomaterials have been proposed so that occupational safety and health regulators properly assess any toxicological concerns and overcome regulatory hurdles 102,125 . ...
... In previous studies, single-walled carbon nanotubes presented lower toxicity due to their ability to cause less lysosomal damage and less secretion of inflammatory factors than multi-walled carbon nanotubes. 52 Although our fabricated Gd-MCNs showed a certain increase in inflammatory factors secretion, it was lower than Gd-DTPA with a clinical dosage. Considering the widely used Gd-DTPA in clinic, the increase in secretion of inflammatory factors by Gd-MCNs was clinically acceptable. ...
Article
Full-text available
Purpose Magnetic resonance imaging (MRI) has been a valuable and widely used examination technique in clinical diagnosis and prognostic efficacy evaluation. The introduction of MRI contrast agent (CA) improves its sensitivity obviously, particularly with the development of nano-CA, which presents higher contrast enhancement ability. However, systematical evaluation of their toxicity is still limited, hampering their further translation in clinics. Methods In this paper, to systematically evaluate the toxicity of nano-CA, Gd-doped mesoporous carbon nanoparticles (Gd-MCNs) prepared by a one-step hard template method were introduced as a model and clinically used MRI CA, Magnevist (Gd-DTPA) as control. Their in vitro blood compatibility, cellular toxicity, DNA damage, oxidative stress, inflammation response as well as in vivo toxicity and MR imaging behaviors were studied and compared. Results The experimental results showed that compared with Gd-DTPA, Gd-MCNs displayed negligible influence on the red blood cell shape, aggregation, BSA structure, macrophage morphology and mitochondrial function. Meanwhile, limited ROS and inflammatory cytokine production also illustrated the cellular compatibility of Gd-MCNs. For in vivo toxicity evaluation, Gd-MCNs presented acceptable in vivo biosafety even under 12 times injection for 12 weeks. More importantly, at the same concentration of Gd, Gd-MCNs displayed better contrast enhancement of tumor than Gd-DTPA, mainly coming from its high MRI relaxation rate which is nearly 9 times that of Gd-DTPA. Conclusion In this paper, we focus on the toxicity evaluation of MRI nano-CA, Gd-MCNs from different angles. With Gd-DTPA as control, Gd-MCNs appeared to be highly biocompatible and safe nanoparticles that possessed promising potentials for the use of MRI nano-CA. In the future, more research on the long-term genotoxicity and the fate of nanoparticles after being swallowed should be performed.
... Based on these observations, carbon nanotube of ~50 nm-diameter was designated as Group 2B (possibly carcinogenic to humans) by IARC in 2014 [80]. Recently, we have identified receptors for carbon nanotubes in murine macrophages as Tim4 [81] and in human macrophages as Siglec-14, which provokes inflammation on activation by carbon nanotube [82]. ...
Article
Full-text available
Current progress in biology and medical science is based on the observation at the level of nanometers via electron microscopy and computation. Of note, the size of most cells in higher species exists in a limited range from 5 to 50 μm. Recently, it was demonstrated that endogenous extracellular nanoparticles play a role in communication among various cellular types in a variety of contexts. Among them, exosomes in serum have been established as biomarkers for human diseases by analyzing the cargo molecules. No life on the earth can survive without iron. However, excess iron can be a risk for carcinogenesis in rodents and humans. Nano-sized molecules may cause unexpected bioeffects, including carcinogenesis, which is a process to establish cellular iron addiction with ferroptosis-resistance. Asbestos and carbon nanotubes are the typical examples, leading to carcinogenesis by the alteration of iron metabolism. Recently, we found that CD63, one of the representative markers of exosomes, is under the regulation of iron-responsive element/iron-regulatory protein system. This is a safe strategy to share excess iron in the form of holo-ferritin between iron-sufficient and -deficient cells. On the other hand, damaged cells may secrete holo-ferritin-loaded exosomes as in the case of macrophages in ferroptosis after asbestos exposure. These holo-ferritin-loaded exosomes can cause mutagenic DNA damage in the recipient mesothelial cells. Thus, there is an iron link between exogenous and endogenous nanoparticles, which requires further investigation for better understanding and the future applications.
... Cell viability e.g., ATP assay 146 , trypan blue cell counting 147 , LDH assay 147,148 Cell proliferation e.g., WST-1 assay 148 Biomarkers for cell death mechanisms e.g., PARP (apoptosis) 149 , RIP (necrosis) 149 , GSDMD (pyroptosis) 150 , Caspase 150 Autophagy activation 151 Genotoxicity γH2AX detection 152,153 Micronucleus assay 154,155 Immune response Proinflammatory cytokine measurement, e.g., IL-1β, IL-4, TNF-α 156,157 Activation of pattern recognition receptor pathways, e.g., NLRP3 inflammasome activation 158 Inflammatory markers, e.g., white blood cell count, neutrophils, lymphocytes, monocytes, eosinophils 65 Immune cell activation 157 Oxidative stress Acellular oxidative potential assay 159 ROS measurement 146,153 Cell/organelle morphology Brightfield microscopy TEM 153,160 Membrane damage by L-leucyl-L-leucin methyl ester 156,161 Absorption, distribution, metabolism, and excretion NIR fluorescence imaging 13 Raman mapping 162 Confocal fluorescence microscopy 163,164 In vivo toxicity Survival 65 Weight change 65 Tissue-or organ-specific damages e.g., serum biomarker for hepatic injury or renal function 65 Blood oxygenation 65 ...
Preprint
Full-text available
Carbon nanotubes are a large family of carbon-based hollow cylindrical structures with unique physicochemical properties that have motivated research for diverse applications; some have reached commercialization. Recent actions in the European Union that propose to ban this entire class of materials highlight an unmet need to precisely define carbon nanotubes, to better understand their toxicological risk effects on human health and the environment throughout their life cycle, and to communicate science-based policy-driving information comprising their taxonomy, safe sourcing, processing, production, manufacturing, handling, use, transportation, and disposal. In this review, we discuss current information and knowledge gaps regarding these issues. We highlight the significance of life cycle assessments of carbon nanotubes and provide a framework to inform policy decisions.
... As a result, an immune-suppressive environment is created that supports tumor growth and invasion (Figure 4). However, despite the regulatory functions of most Siglecs in suppressing inflammation and modulating immune suppression, three specific Siglecs (Siglec-14, 15, and 16) have been identified to incite inflammation and stimulate immune activation [148][149][150]. The intricate mechanism underlying the interaction between sialylation and Siglecs in the tumor microenvironment necessitates further exploration. ...
Article
Full-text available
The tumor microenvironment (TME), where the tumor cells incite the surrounding normal cells to create an immune suppressive environment, reduces the effectiveness of immune responses during cancer development. Sialylation, a type of glycosylation that occurs on cell surface proteins, lipids, and glycoRNAs, is known to accumulate in tumors and acts as a “cloak” to help tumor cells evade immunological surveillance. In the last few years, the role of sialylation in tumor proliferation and metastasis has become increasingly evident. With the advent of single-cell and spatial sequencing technologies, more research is being conducted to understand the effects of sialylation on immunity regulation. This review provides updated insights into recent research on the function of sialylation in tumor biology and summarizes the latest developments in sialylation-targeted tumor therapeutics, including antibody-mediated and metabolic-based sialylation inhibition, as well as interference with sialic acid–Siglec interaction.
... 670 Similarly, the MD simulation identified a novel CNT-receptor binding mode mediated by multiple aromatic−aromatic interactions. 671 Recently, a CNN deep learning model was successfully trained and employed to assist single-vessel analysis of NP permeability in tumor vasculatures. 672 Using NP library and computational methods (i.e., molecular docking and ML), two novel broad-spectrum adjuvants were identified and experimentally validated. ...
Article
Decades of nanotoxicology research have generated extensive and diverse data sets. However, data is not equal to information. The question is how to extract critical information buried in vast data streams. Here we show that artificial intelligence (AI) and molecular simulation play key roles in transforming nanotoxicity data into critical information, i.e., constructing the quantitative nanostructure (physicochemical properties)-toxicity relationships, and elucidating the toxicity-related molecular mechanisms. For AI and molecular simulation to realize their full impacts in this mission, several obstacles must be overcome. These include the paucity of high-quality nanomaterials (NMs) and standardized nanotoxicity data, the lack of model-friendly databases, the scarcity of specific and universal nanodescriptors, and the inability to simulate NMs at realistic spatial and temporal scales. This review provides a comprehensive and representative, but not exhaustive, summary of the current capability gaps and tools required to fill these formidable gaps. Specifically, we discuss the applications of AI and molecular simulation, which can address the large-scale data challenge for nanotoxicology research. The need for model-friendly nanotoxicity databases, powerful nanodescriptors, new modeling approaches, molecular mechanism analysis, and design of the next-generation NMs are also critically discussed. Finally, we provide a perspective on future trends and challenges.