Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Colloid Interface Sci ; 664: 766-778, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38492378

ABSTRACT

Developing multi-functional electrocatalysts is of great practical significance for fuel cells and water splitting. Herein, Rh-Rh2O3 nanoclusters are prepared and the surface oxygen vacancy content is regulated elaborately by post-treatment. The optimized Rh-Rh2O3/C-400 exhibits superior trifunctional catalytic activity for hydrogen oxidation reaction (HOR), hydrogen evolution reaction (HER) and hydrazine oxidation reaction (HzOR), i.e., the mass activity for HOR is 2.29 mA µgRh-1, and the overpotential for HER and HzOR at 10 mA cm-2 is as low as 12 mV and 31 mV, respectively, superior to the benchmark Pt/C. Rh-Rh2O3/C-400 also displays promising performance in practical devices, with the H2-O2 anion-exchange-membrane fuel cell delivering a peak power density of 0.66 W cm-2, and the hydrazine-assisted water splitting electrolyzer requiring a low electrolysis voltage of 0.161 V at 0.1 A cm-2. The experimental and theoretical investigations discover that the hydrogen binding energy (HBE) is linearly depended on surface oxygen vacancy contents, and the HBE directly determines the catalytic activity for HOR, HER and HzOR. This work not only innovates an efficient Rh-based nanocluster tri-functional electrocatalyst, but also eludicates the intrinsic relationship of surface structure-intermediate adsorption-catalytic activity.

2.
Nat Commun ; 14(1): 5523, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37684225

ABSTRACT

Nonlinear optics provides a means to bridge between different electromagnetic frequencies, enabling communication between visible, infrared, and terahertz bands through χ(2) and higher-order nonlinear optical processes. However, precisely modulating nonlinear optical waves in 3D space remains a significant challenge, severely limiting the ability to directly manipulate optical information across different wavelength bands. Here, we propose and experimentally demonstrate a three-dimensional (3D) χ(2)-super-pixel hologram with nanometer resolution in lithium niobate crystals, capable of performing advanced processing tasks. In our design, each pixel consists of properly arranged nanodomain structures capable of completely and dynamically manipulating the complex-amplitude of nonlinear waves. Fabricated by femtosecond laser writing, the nonlinear hologram features a pixel diameter of 500 nm and a pixel density of approximately 25000 pixels-per-inch (PPI), reaching far beyond the state of the art. In our experiments, we successfully demonstrate the novel functions of the hologram to process near-infrared (NIR) information at visible wavelengths, including dynamic 3D nonlinear holographic imaging and frequency-up-converted image recognition. Our scheme provides a promising nano-optic platform for high-capacity optical storage and multi-functional information processing across different wavelength ranges.

3.
Nanoscale ; 15(31): 12868-12879, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37492026

ABSTRACT

A growing number of nanomaterials are being broadly used in food-related fields as well as therapeutics. Oral exposure to these widespread nanomaterials is inevitable, with the intestine being a major target organ. Upon encountering the intestine, these nanoparticles can cross the intestinal barrier, either bypassing cells or via endocytosis pathways to enter the adjacent mesentery. The intricate structure of the mesentery and its entanglement with the abdominal digestive organs determine the final fate of nanomaterials in the human body. Importantly, mesentery-governed dynamic processes determine the distribution and subsequent biological effects of nanomaterials that cross the intestine, thus there is a need to understand how nanomaterials interact with the mesentery. This review presents the recent progress in understanding the mesenteric structure and function and highlights the importance of the mesentery in health and disease, with a focus on providing new insights and research directions around the biological effects of nanomaterials on the mesentery. A thorough comprehension of the interactions between nanomaterials and the mesentery will facilitate the design of safer nanomaterial-containing products and the development of more effective nanomedicines to combat intestinal disorders.


Subject(s)
Nanoparticles , Nanostructures , Humans , Mesentery/metabolism , Nanoparticles/chemistry
4.
Sci Adv ; 9(27): eadg2252, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37418525

ABSTRACT

The blood circulation is considered the only way for the orally administered nanoparticles to enter the central nervous systems (CNS), whereas non-blood route-mediated nanoparticle translocation between organs is poorly understood. Here, we show that peripheral nerve fibers act as direct conduits for silver nanomaterials (Ag NMs) translocation from the gut to the CNS in both mice and rhesus monkeys. After oral gavage, Ag NMs are significantly enriched in the brain and spinal cord of mice with particle state however do not efficiently enter the blood. Using truncal vagotomy and selective posterior rhizotomy, we unravel that the vagus and spinal nerves mediate the transneuronal translocation of Ag NMs from the gut to the brain and spinal cord, respectively. Single-cell mass cytometry analysis revealed that enterocytes and enteric nerve cells take up significant levels of Ag NMs for subsequent transfer to the connected peripheral nerves. Our findings demonstrate nanoparticle transfer along a previously undocumented gut-CNS axis mediated by peripheral nerves.


Subject(s)
Nanostructures , Silver , Animals , Mice , Central Nervous System , Spinal Cord , Peripheral Nerves
5.
Environ Sci Technol ; 57(21): 7891-7901, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37163641

ABSTRACT

Exposure to atmospheric particulate matter (PM) is a frequent occurrence to humans, and their adverse outcomes have become a global concern. Although PM-induced inflammation is a common phenomenon, a clear picture of the mechanisms underlying exosome-mediated inflammation of PM has not yet emerged. Here, we show that exosomes can mediate the cascade reactions for the transfer of PM and inflammatory responses of macrophages. Specifically, two fine PM2.5, namely F1 (<0.49 µm) and F2 (0.95-1.5 µm), stimulated a substantial release of exosomes from macrophages (THP-1 cells) with the order of F1 > F2, via regulation of the P2X7 receptor (P2X7R). Inhibiting P2X7R with a specific inhibitor largely prevented the secretion of exosomes. In particular, we found that exosomes served as a mediator for the transfer of PM2.5 to the recipient macrophages and activated NF-κB signaling through toll-like receptor 4 (TLR-4), thereby stimulating inflammatory cytokine release and altering the inflammatory phenotype of recipients. Importantly, the exosomes derived from PM2.5-treated macrophages induced the inflammatory responses of lung in mice. Our results highlight that exosomes undergo a secretion-particle transfer-adverse outcome chain in macrophages treated with PM2.5. Given the ubiquitous atmospheric particulate matter, these new findings underscore an urgent need for assessing the secretion of exosomes and their impact on human health via exosome-centric physiological pathways.


Subject(s)
Exosomes , Particulate Matter , Mice , Humans , Animals , Particulate Matter/toxicity , Exosomes/metabolism , Cytokines/metabolism , Macrophages/metabolism , Inflammation/chemically induced , Inflammation/metabolism
6.
Proc Natl Acad Sci U S A ; 120(20): e2218739120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155879

ABSTRACT

Carbon-based nanomaterials (CNMs) have recently been found in humans raising a great concern over their adverse roles in the hosts. However, our knowledge of the in vivo behavior and fate of CNMs, especially their biological processes elicited by the gut microbiota, remains poor. Here, we uncovered the integration of CNMs (single-walled carbon nanotubes and graphene oxide) into the endogenous carbon flow through degradation and fermentation, mediated by the gut microbiota of mice using isotope tracing and gene sequencing. As a newly available carbon source for the gut microbiota, microbial fermentation leads to the incorporation of inorganic carbon from the CNMs into organic butyrate through the pyruvate pathway. Furthermore, the butyrate-producing bacteria are identified to show a preference for the CNMs as their favorable source, and excessive butyrate derived from microbial CNMs fermentation further impacts on the function (proliferation and differentiation) of intestinal stem cells in mouse and intestinal organoid models. Collectively, our results unlock the unknown fermentation processes of CNMs in the gut of hosts and underscore an urgent need for assessing the transformation of CNMs and their health risk via the gut-centric physiological and anatomical pathways.


Subject(s)
Gastrointestinal Microbiome , Nanostructures , Nanotubes, Carbon , Humans , Animals , Mice , Gastrointestinal Microbiome/physiology , Nanotubes, Carbon/adverse effects , Fermentation , Butyrates/metabolism
7.
J Mater Chem B ; 11(9): 1904-1915, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36734837

ABSTRACT

Due to their excellent antibacterial ability, silver nanomaterials (Ag NMs) are the most frequently used nanomaterials. Their widespread use introduces the risk of human ingestion. However, the potential toxicity of Ag NMs to the gut microbiota and their metabolic profile are yet to be fully explored. In this study, we examined the effects of Ag NMs after oral administration (0.5 mg kg-1 and 2.5 mg kg-1, 14 and 28 days) on gut homeostasis by integrating tissue imaging, 16s rRNA gene sequencing and metabolomics techniques. We uncovered that silver nanoparticles (Ag NPs) and silver nanowires (Ag NWs) altered the structure (inhibiting the proliferation of Gram-negative bacteria) and decreased the diversity of gut microbiota in mice after short-term (14 days) exposure, while the microbial community tended to recover after long-term exposure (28 days), indicating that the resistance and resilience of the gut microbiome may pose a defense against the interference by reactive, exogenous nanomaterials. Interestingly, even though the gut microbiota structure recovered after 28 days of exposure, the gut metabolites significantly changed, showing increased 1H-indole-3-carboxylic acid and elevated levels of 5-HT in the gut and blood. Collectively, our results provide a piece of evidence on the association between the ingestion of exogenous nanoparticles and gut homeostasis, especially the metabolic profile of the host. This work thus provides additional insights for the continued investigation of the adverse effects of silver nanomaterials on biological hosts.


Subject(s)
Gastrointestinal Microbiome , Metal Nanoparticles , Nanostructures , Nanowires , Mice , Humans , Animals , Gastrointestinal Microbiome/genetics , Silver/chemistry , Serotonin , Metal Nanoparticles/chemistry , RNA, Ribosomal, 16S/genetics , Metabolome , Administration, Oral
8.
Nano Res ; 16(3): 3976-3990, 2023.
Article in English | MEDLINE | ID: mdl-36465523

ABSTRACT

With research burgeoning in nanoscience and nanotechnology, there is an urgent need to develop new biological models that can simulate native structure, function, and genetic properties of tissues to evaluate the adverse or beneficial effects of nanomaterials on a host. Among the current biological models, three-dimensional (3D) organoids have developed as powerful tools in the study of nanomaterial-biology (nano-bio) interactions, since these models can overcome many of the limitations of cell and animal models. A deep understanding of organoid techniques will facilitate the development of more efficient nanomedicines and further the fields of tissue engineering and personalized medicine. Herein, we summarize the recent progress in intestinal organoids culture systems with a focus on our understanding of the nature and influencing factors of intestinal organoid growth. We also discuss biomimetic extracellular matrices (ECMs) coupled with nanotechnology. In particular, we analyze the application prospects for intestinal organoids in investigating nano-intestine interactions. By integrating nanotechnology and organoid technology, this recently developed model will fill the gaps left due to the deficiencies of traditional cell and animal models, thus accelerating both our understanding of intestine-related nanotoxicity and the development of nanomedicines.

9.
Dalton Trans ; 51(23): 9167-9174, 2022 Jun 13.
Article in English | MEDLINE | ID: mdl-35670450

ABSTRACT

Support corrosion is a traditional intractable problem for oxygen electrodes of fuel cells, so developing anti-corrosion supports is highly desirable. Herein, we fabricate a three-dimensional (3D) interconnected-graphene enveloped titanium dioxide flower (TiO2@RGO) as a robust support for the oxygen reduction reaction (ORR). Benefiting from the unique 3D architecture, the TiO2@RGO composite possesses both a large surface area of 174 m2 g-1 and a superior electrical conductivity of 0.19 S cm-1, enabling an electron highway for efficient simultaneous mass transfer of reactants. After loading Pt nanoparticles, the Pt-TiO2@RGO catalyst exhibits a similar catalytic activity to the commercial Pt/C catalyst, while superior stability under the accelerated degradation protocols for both catalysts (0.6-1.0VRHE) and supports (1.0-1.5VRHE), due to the strong metal-support interaction (SMSI) of Pt nanoparticles and the TiO2@RGO composite support. The PEMFC with the Pt-TiO2@RGO cathode delivers a peak power density of 901 mW cm-2, which is comparable to that with a Pt/C cathode. This work proposes a new strategy for designing robust catalyst supports for PEMFCs.

10.
ACS Appl Mater Interfaces ; 14(8): 10246-10256, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35184551

ABSTRACT

Designing high-performance hydrogen evolution reaction (HER) catalysts is crucial for seawater splitting. Herein, we demonstrate a facile Anderson-type polyoxometalate-assisted synthesis route to prepare defect-rich doped 1T/2H-MoSe2 nanosheets. As demonstrated, the optimized defect-rich doped 1T/2H-MoSe2 nanosheets display low overpotentials of 116 and 274 mV to gain 10 mA cm-2 in acidic and simulated seawater for the HER, respectively. A magnesium (Mg)/seawater battery was fabricated with the defect-rich doped 1T/2H-MoSe2 nanosheet cathode, displaying the highest power density of up to 7.69 mW cm-2 and stable galvanostatic discharging over 24 h. The theoretical and experimental investigations show that the superior HER and battery performances of the heteroatom-doped MoSe2 nanosheets are attributed to both the improved intrinsic catalytic activity (effective activation of water and favorable subsequent hydrogen desorption) and the abundant active sites, benefiting from the favorable catalytic factors of the doped heteroatom, 1T phase, and defects. Our work presents an intriguing structural modulation strategy to design high-performance catalysts toward both HER and Mg/seawater batteries.

11.
ACS Nano ; 15(10): 15858-15873, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34622660

ABSTRACT

The biological effect of engineered carbon nanotubes (CNTs) as beneficial biomaterials on the intestine, especially on its development, remains unclear. Here, we investigated the profitable effect of CNTs with a different graphene layer and surface modification on the 3D model of intestinal organoids and demonstrated that CNTs (50 µg/mL) promoted the development of intestinal organoids over time (0-5 days). The mechanisms involve the modulation of extracellular matrix (ECM) viscoelasticity and intracellular energy metabolism. In particular, CNTs reduced the hardness of the extracellular matrix through decreasing the elasticity and increasing the viscosity as a result of elevated metalloproteinase and binding to a protein scaffold, which activated the mechanical membrane sensors of cells, Piezo, and downstream P-p38-yes-associated protein (YAP) pathway. Moreover, CNTs altered the metabolic profile of intestinal organoids and induced increased mitochondria activity, respiration, and nutrient absorption. These mechanisms cooperated with each other to promote the proliferation and differentiation of intestinal organoids. In addition, the promoted effect of CNTs is highly dependent on the number of graphene layers, manifested as multiwalled CNTs > single-walled CNTs. Our findings highlight the CNT-intestine interaction and imply the potential of CNTs as biomaterials for intestine-associated tissue engineering.


Subject(s)
Nanotubes, Carbon , Energy Metabolism , Extracellular Matrix/metabolism , Intestines , Organoids , Viscosity
12.
Nanoscale ; 13(19): 8806-8816, 2021 May 20.
Article in English | MEDLINE | ID: mdl-33904557

ABSTRACT

The widespread occurrence of microplastics (MPLs) and nanoplastics (NPLs), collectively abbreviated as M/NPLs, has markedly affected the ecosystem and has become a global threat to human health. Multiple investigations have shown that the chronic ingestion of M/NPLs negatively affects gut barrier function but the mechanism remains unclear. Herein, this research has investigated the toxic effects of pristine polystyrene (PS) M/NPLs, negatively charged carboxylated polystyrene M/NPLs (PS-COOH) and positively charged aminated polystyrene M/NPLs (PS-NH2) of two sizes (70 nm and 5 µm in diameter) in mice. Gavage of these PS M/NPLs for 28 days caused obvious injuries to the gut tract, leading to the decreased expression of tight junction proteins. The toxicity of the M/NPLs was ranked as PS-NH2 > PS-COOH > pristine PS. Oral administration of these M/NPLs resulted in marked gut microbiota dysbiosis. The M/NPLs-enriched genera generally contained opportunistic pathogens which are accompanied by a deteriorated intestinal barrier function, while most M/NPLs-decreased bacteria were beneficial microbes with known tight junction-promoting functions, implicating an important indirect toxic effect of gut microbiota dysbiosis in M/NPLs-induced gut barrier dysfunction. In conclusion, this research highlights the importance of gut microbiota in the toxicity of M/NPLs exposure on gut barrier function, providing novel insights into the adverse effects of M/NPLs exposure on human health.


Subject(s)
Gastrointestinal Microbiome , Microplastics , Animals , Dysbiosis , Ecosystem , Mice , Plastics
13.
ACS Nano ; 14(11): 14391-14416, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33138351

ABSTRACT

Recent clinical and pathological evidence have implicated the gut microbiota as a nexus for modulating the homeostasis of the human body, impacting conditions from cancer and dementia to obesity and social behavior. The connections between microbiota and human diseases offer numerous opportunities in medicine, most of which have limited or no therapeutic solutions available. In light of this paradigm-setting trend in science, this review aims to provide a comprehensive and timely summary of the mechanistic pathways governing the gut microbiota and their implications for nanomedicines targeting cancer and neurodegenerative diseases. Specifically, we discuss in parallel the beneficial and pathogenic relationship of the gut microbiota along the gut-brain and gut-cancer axes, elaborate on the impact of dysbiosis and the gastrointestinal corona on the efficacy of nanomedicines, and highlight a molecular mimicry that manipulates the universal cross-ß backbone of bacterial amyloid to accelerate neurological disorders. This review further offers a forward-looking section on the rational design of cancer and dementia nanomedicines exploiting the gut-brain and gut-cancer axes.


Subject(s)
Gastrointestinal Microbiome , Nanomedicine , Neoplasms , Neurodegenerative Diseases , Brain , Humans
14.
Langmuir ; 36(33): 9800-9809, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32787117

ABSTRACT

Because of their distinctive mode of action in targeting bacterial cell membranes, antimicrobial peptides (AMPs) are increasingly regarded as a potential candidate for the development of novel antibiotics to combat the wide spread of bacterial resistance. To date, understanding of the exact molecular process by which AMPs act on the real bacterial envelope remains challenging. Simultaneously, the aggregated state of AMPs upon interaction with bacterial envelopes is still elusive. Previously, we have demonstrated that the potent antibacterial activity of a designed surfactant-like peptide Ac-A9K-NH2 benefited greatly from its high self-assembling ability and appropriate self-assembled morphologies and sizes. By using high-resolution atomic force microscopy, we here not only follow the variations of the Escherichia coli cell envelope in the presence of Ac-A9K-NH2 but also characterize the peptide aggregates on the bacterial surface as well as on the substrate surface. The results, together with those from fluorescence, zeta potential, circular dichroism, and scanning electron microscopy measurements, indicate that both the positively charged peptide monomers and self-assembled nanostructures can directly act on the negatively charged bacterial surface, followed by their insertion into the bacterial membrane, the formation of surface nanopores, and membrane lysis. The mechanism of Ac-A9K-NH2 against E. coli is thus consistent with the detergent-like mode of action. This work enhances our mechanistic understanding of the antibacterial behaviors of self-assembling peptides that will be valuable in exploring their biomedical applications.


Subject(s)
Antimicrobial Cationic Peptides , Escherichia coli , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Membrane , Circular Dichroism
15.
Sci China Life Sci ; 63(8): 1168-1182, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32458255

ABSTRACT

Due to their many advantageous properties, nanomaterials (NMs) have been utilized in diverse consumer goods, industrial products, and for therapeutic purposes. This situation leads to a constant risk of exposure and uptake by the human body, which are highly dependent on nanomaterial size. Consequently, an improved understanding of the interactions between different sizes of nanomaterials and biological systems is needed to design safer and more clinically relevant nano systems. We discuss the sizedependent effects of nanomaterials in living organisms. Upon entry into biological systems, nanomaterials can translocate biological barriers, distribute to various tissues and elicit different toxic effects on organs, based on their size and location. The association of nanomaterial size with physiological structures within organs determines the site of accumulation of nanoparticles. In general, nanomaterials smaller than 20 nm tend to accumulate in the kidney while nanomaterials between 20 and 100 nm preferentially deposit in the liver. After accumulating in organs, nanomaterials can induce inflammation, damage structural integrity and ultimately result in organ dysfunction, which helps better understand the size-dependent dynamic processes and toxicity of nanomaterials in organisms. The enhanced permeability and retention effect of nanomaterials and the utility of this phenomenon in tumor therapy are also highlighted.


Subject(s)
Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Animals , Antineoplastic Agents/adverse effects , Biocompatible Materials/adverse effects , Biological Transport , Drug Carriers/adverse effects , Humans , Inflammation/chemically induced , Kidney/metabolism , Kinetics , Liver/metabolism , Lung/metabolism , Metals/metabolism , Nanoparticles/adverse effects , Oxides/metabolism , Particle Size , Structure-Activity Relationship , Surface Properties , Tissue Distribution/drug effects
16.
Chem Res Toxicol ; 33(5): 1082-1109, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32302095

ABSTRACT

The interplay between nanotechnology and pathogens offers a new quest to fight against human infections. Inspiring from their unique thermal, magnetic, optical, or redox potentials, numerous nanomaterials have been employed for bacterial theranostics. The past decade has seen dramatic progress in the development of various nanoantimicrobials, which demands more focus on their safety assessment. The present review critically discusses the toxicity of nanoantimicrobials and the role of key features, including composition, size, surface charge, loading capability, hydrophobicity/philicity, precise release, and functionalization, that can contribute to modulating the effects on microbes. Moreover, how differences in microbe's structure, biofilm formation, persistence cells, and intracellular pathogens bestow resistance or sensitivity toward nanoantimicrobials is broadly investigated. In extension, the most important types of nanoantimicrobial with clinical prospective and their safety assessment are summarized, and finally, based on available evidence, an insight of the principles in designing safer nanoantimicrobials for overcoming pathogens and future challenges in the field is provided.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bacteria/drug effects , Nanostructures/adverse effects , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Nanostructures/chemistry , Surface Properties
17.
Small ; 16(21): e1907665, 2020 05.
Article in English | MEDLINE | ID: mdl-32347646

ABSTRACT

Engineered nanomaterials (ENMs) are used in food additives, food packages, and therapeutic purposes owing to their useful properties, Therefore, human beings are orally exposed to exogenous nanomaterials frequently, which means the intestine is one of the primary targets of nanomaterials. Consequently, it is of great importance to understand the interaction between nanomaterials and the intestine. When nanomaterials enter into gut lumen, they inevitably interact with various components and thereby display different effects on the intestine based on their locations; these are known as location-oriented effects (LOE). The intestinal LOE confer a new biological-effect profile for nanomaterials, which is dependent on the involvement of the following biological processes: nano-mucus interaction, nano-intestinal epithelial cells (IECs) interaction, nano-immune interaction, and nano-microbiota interaction. A deep understanding of NM-induced LOE will facilitate the design of safer NMs and the development of more efficient nanomedicine for intestine-related diseases. Herein, recent progress in this field is reviewed in order to better understand the LOE of nanomaterials. The distant effects of nanomaterials coupling with microbiota are also highlighted. Investigation of the interaction of nanomaterials with the intestine will stimulate other new research areas beyond intestinal nanotoxicity.


Subject(s)
Intestines , Nanostructures , Humans , Intestines/drug effects , Nanostructures/toxicity
19.
Toxicol Sci ; 172(2): 398-410, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31504961

ABSTRACT

Carbon nanomaterials (CNMs) are widely used in industrial and medical sectors. The increasing exposure of CNMs necessitates the studies of their potential environmental and health effects. High-mobility group box-1 (HMGB1) is a nuclear DNA-binding protein, but when released from cells, may cause sustained inflammatory response and promote cell migration and invasion. In this work, we found that 7-day exposure of 2.5 mg/kg/day CNMs, including C60, single-walled carbon nanotubes, and graphene oxides significantly elevated the level of HMGB1 in blood and lung lavage fluids in C57BL/6 mice. Subsequently, cellular effects and underlying mechanism were explored by using Raw264.7. The results showed that noncytotoxic CNMs enhanced HMGB1 intracellular translocation and release via activating P2X7 receptor. Released HMGB1 further activated receptor for advanced glycation endproducts (RAGE) and downstream signaling pathway by upregulating RAGE and Rac1 expression. Simultaneously, CNMs prepared the cells for migration and invasion by modulating MMP2 and TIMP2 gene expression as well as cytoskeleton reorganization. Intriguingly, released HMGB1 from macrophages promoted the migration of nearby lung cancer cell, which can be efficiently inhibited by neutralizing antibodies against HMGB1 and RAGE. Taken together, our work demonstrated that CNMs stimulated HMGB1 release and cell migration/invasion through P2X7R-HMGB1-RAGE pathway. The revealed mechanisms might facilitate a better understanding on the inflammatory property and subsequent cell functional alteration of CNMs.


Subject(s)
Cell Movement/drug effects , Fullerenes/toxicity , Graphite/toxicity , HMGB1 Protein/blood , Macrophages/drug effects , Nanotubes, Carbon/toxicity , A549 Cells , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Survival/drug effects , Coculture Techniques , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred C57BL , Protein Transport , RAW 264.7 Cells , Receptor for Advanced Glycation End Products/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction
20.
Sci Adv ; 5(9): eaax0937, 2019 09.
Article in English | MEDLINE | ID: mdl-31523712

ABSTRACT

Carbonic anhydrase (CA) IX overexpresses exclusively on cell membranes of hypoxic tumors, regulating the acidic tumor microenvironment. Small molecules of CA inhibitor modified with short peptide successfully achieve CA IX-targeted self-assembly that localizes CA inhibitors on hypoxic cancer cell surfaces and enhances their inhibition efficacy and selectivity. CA IX-related endocytosis also promotes selective intracellular uptake of these nanofibers under hypoxia, in which nanofiber structures increase in size with decreasing pH. This effect subsequently causes intracellular acid vesicle damage and blocks protective autophagy. The versatility of tunable nanostructures responding to cell milieu impressively provokes selective toxicities and provides strategic therapy for hypoxic tumors. Moreover, in vivo tests demonstrate considerable antimetastatic and antiangiogenesis effects in breast tumors, and particularly remarkable enhancement of antitumor efficacy in doxorubicin administration. With its biocompatible components and distinctive hypoxia therapies, this nanomaterial advances current chemotherapy, providing a new direction for hypoxic cancer therapy.


Subject(s)
Breast Neoplasms , Carbonic Anhydrase Inhibitors , Doxorubicin , Nanofibers , Peptides , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Humans , Mice , Mice, Nude , Nanofibers/chemistry , Nanofibers/therapeutic use , Peptides/chemistry , Peptides/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...