Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 240
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; : e2310540, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38597766

RESUMEN

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.

2.
Nanomedicine ; 55: 102714, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38738528

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with poor survival rates. Here, we evaluated iron-doped hydroxyapatite (FeHA) as a potential nanomedicine-based approach to combat PDAC. FeHA, in combination with a sublethal dose of the glutathione peroxidase 4 (GPX4) inhibitor RSL3, was found to trigger ferroptosis in KRAS mutant PANC-1 cells, but not in BxPC3 cells, while sparing normal human cells (fibroblasts and peripheral blood mononuclear cells). These findings were recapitulated in 3D spheroids generated using PDAC cells harboring wild-type versus mutant KRAS. Moreover, ferroptosis induction by FeHA plus RSL3 was reversed by the knockdown of STEAP3, a metalloreductase responsible for converting Fe3+ to Fe2+. Taken together, our data show that FeHA is capable of triggering cancer cell death in a KRAS-selective, STEAP3-dependent manner in PDAC cells.


Asunto(s)
Carcinoma Ductal Pancreático , Ferroptosis , Hierro , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Hierro/química , Hierro/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Ferroptosis/efectos de los fármacos , Línea Celular Tumoral , Nanopartículas/química , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
3.
Biochem Biophys Res Commun ; 633: 45-51, 2022 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-36344160

RESUMEN

Nanomaterials are marvelously small, yet they may be harnessed to great effect. Much has been learned with respect to the biological interactions and effects of nanomaterials during the past decade. However, characterization of nanomaterials is typically performed on as-synthesized materials. We posit that nanomaterials are dynamic entities and should be studied and regulated as such. Hence, characterization of nanomaterials should take into account their biotransformation. However, in situ characterization of nanomaterials as they undergo dynamic changes (coronation, dissolution, degradation) in a living system remains a formidable challenge in nanosafety. Material scientists and toxicologists need to join forces to address this issue.


Asunto(s)
Nanoestructuras , Nanoestructuras/toxicidad
4.
Small ; 18(20): e2107816, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35434920

RESUMEN

Trained immunity is a recently described phenomenon whereby cells of the innate immune system undergo long-term epigenetic and/or metabolic reprogramming following a short-term interaction with microbes or microbial products. Here, it is shown that 2D transition metal dichalcogenides (TMDs) trigger trained immunity in primary human monocyte-derived macrophages. First, aqueous dispersions of 2D crystal formulations of MoS2 and WS2 are tested, and no cytotoxicity is found despite avid uptake of these materials by macrophages. However, when macrophages are pre-exposed to TMDs, followed by a resting period, this causes a marked modulation of immune-specific gene expression upon subsequent challenge with a microbial agent (i.e., bacterial lipopolysaccharides). Specifically, MoS2 triggers trained immunity through an epigenetic pathway insofar as the histone methyltransferase inhibitor methylthioadenosine reverses these effects. Furthermore, MoS2 triggers an elevation of cyclic adenosine monophosphate (cAMP) levels in macrophages and increased glycolysis is also evidenced in cells subjected to MoS2 training, pointing toward a metabolic rewiring of the cells. Importantly, it is observed that MoS2 triggers the upregulation of Mo-dependent enzymes in macrophages, thus confirming that Mo is bioavailable in these cells. In conclusion, MoS2 is identified as a novel inducer of trained immunity. Thus, TMDs could potentially be harnessed as immunomodulatory agents.


Asunto(s)
Inmunidad Innata , Molibdeno , Epigénesis Genética , Humanos , Macrófagos/metabolismo , Redes y Vías Metabólicas , Molibdeno/farmacología
5.
Part Fibre Toxicol ; 19(1): 33, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35538581

RESUMEN

BACKGROUND: Copper oxide (CuO) nanoparticles (NPs) are known to trigger cytotoxicity in a variety of cell models, but the mechanism of cell death remains unknown. Here we addressed the mechanism of cytotoxicity in macrophages exposed to CuO NPs versus copper chloride (CuCl2). METHODS: The mouse macrophage cell line RAW264.7 was used as an in vitro model. Particle uptake and the cellular dose of Cu were investigated by transmission electron microscopy (TEM) and inductively coupled plasma mass spectrometry (ICP-MS), respectively. The deposition of Cu in lysosomes isolated from macrophages was also determined by ICP-MS. Cell viability (metabolic activity) was assessed using the Alamar Blue assay, and oxidative stress was monitored by a variety of methods including a luminescence-based assay for cellular glutathione (GSH), and flow cytometry-based detection of mitochondrial superoxide and mitochondrial membrane potential. Protein aggregation was determined by confocal microscopy using an aggresome-specific dye and protein misfolding was determined by circular dichroism (CD) spectroscopy. Lastly, proteasome activity was investigated using a fluorometric assay. RESULTS: We observed rapid cellular uptake of CuO NPs in macrophages with deposition in lysosomes. CuO NP-elicited cell death was characterized by mitochondrial swelling with signs of oxidative stress including the production of mitochondrial superoxide and cellular depletion of GSH. We also observed a dose-dependent accumulation of polyubiquitinated proteins and loss of proteasomal function in CuO NP-exposed cells, and we could demonstrate misfolding and mitochondrial translocation of superoxide dismutase 1 (SOD1), a Cu/Zn-dependent enzyme that plays a pivotal role in the defense against oxidative stress. The chelation of copper ions using tetrathiomolybdate (TTM) prevented cell death whereas inhibition of the cellular SOD1 chaperone aggravated toxicity. Moreover, CuO NP-triggered cell death was insensitive to the pan-caspase inhibitor, zVAD-fmk, and to wortmannin, an inhibitor of autophagy, implying that this was a non-apoptotic cell death. ZnO NPs, on the other hand, triggered autophagic cell death. CONCLUSIONS: CuO NPs undergo dissolution in lysosomes leading to copper-dependent macrophage cell death characterized by protein misfolding and proteasomal insufficiency. Specifically, we present novel evidence for Cu-induced SOD1 misfolding which accords with the pronounced oxidative stress observed in CuO NP-exposed macrophages. These results are relevant for our understanding of the consequences of inadvertent human exposure to CuO NPs.


Asunto(s)
Macrófagos , Nanopartículas del Metal , Nanopartículas , Superóxido Dismutasa-1 , Animales , Muerte Celular/efectos de los fármacos , Cobre , Glutatión/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Nanopartículas del Metal/toxicidad , Ratones , Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/toxicidad , Estrés Oxidativo , Pliegue de Proteína/efectos de los fármacos , Células RAW 264.7 , Superóxido Dismutasa-1/metabolismo , Superóxidos
6.
Br J Haematol ; 192(1): 200-211, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33206996

RESUMEN

Severe congenital neutropenia (SCN) of autosomal recessive inheritance, also known as Kostmann disease, is characterised by a lack of neutrophils and a propensity for life-threatening infections. Using whole-exome sequencing, we identified homozygous JAGN1 mutations (p.Gly14Ser and p.Glu21Asp) in three patients with Kostmann-like SCN, thus confirming the recent attribution of JAGN1 mutations to SCN. Using the human promyelocytic cell line HL-60 as a model, we found that overexpression of patient-derived JAGN1 mutants, but not silencing of JAGN1, augmented cell death in response to the pro-apoptotic stimuli, etoposide, staurosporine, and thapsigargin. Furthermore, cells expressing mutant JAGN1 were remarkably susceptible to agonists that normally trigger degranulation and succumbed to a calcium-dependent cell death programme. This mode of cell death was completely prevented by pharmacological inhibition of calpain but unaffected by caspase inhibition. In conclusion, our results confirmed the association between JAGN1 mutations and SCN and showed that SCN-associated JAGN1 mutations unleash a calcium- and calpain-dependent cell death in myeloid cells.


Asunto(s)
Calpaína/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Proteínas de la Membrana/genética , Células Mieloides/metabolismo , Neutropenia/congénito , Apoptosis , Calcio/metabolismo , Muerte Celular , Síndromes Congénitos de Insuficiencia de la Médula Ósea/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Células HL-60 , Humanos , Proteínas de la Membrana/metabolismo , Células Mieloides/citología , Células Mieloides/patología , Neutropenia/genética , Neutropenia/metabolismo , Neutropenia/patología , Mutación Puntual
7.
J Cell Sci ; 132(5)2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30683797

RESUMEN

The Caenorhabditis elegans aminophospholipid translocase TAT-1 maintains phosphatidylserine (PS) asymmetry in the plasma membrane and regulates endocytic transport. Despite these important functions, the structure-function relationship of this protein is poorly understood. Taking advantage of the tat-1 mutations identified by the C. elegans million mutation project, we investigated the effects of 16 single amino acid substitutions on the two functions of the TAT-1 protein. Two substitutions that alter a highly conserved PISL motif in the fourth transmembrane domain and a highly conserved DKTGT phosphorylation motif, respectively, disrupt both functions of TAT-1, leading to a vesicular gut defect and ectopic PS exposure on the cell surface, whereas most other substitutions across the TAT-1 protein, often predicted to be deleterious by bioinformatics programs, do not affect the functions of TAT-1. These results provide in vivo evidence for the importance of the PISL and DKTGT motifs in P4-type ATPases and improve our understanding of the structure-function relationship of TAT-1. Our study also provides an example of how the C. elegans million mutation project helps decipher the structure, functions, and mechanisms of action of important genes.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Intestinos/fisiología , Proteínas de Transferencia de Fosfolípidos/metabolismo , Secuencias de Aminoácidos/genética , Animales , Animales Modificados Genéticamente , Proteínas de Caenorhabditis elegans/genética , Biología Computacional , Endocitosis , Mutación/genética , Fenotipo , Proteínas de Transferencia de Fosfolípidos/genética , Conformación Proteica , Transporte de Proteínas , Relación Estructura-Actividad
8.
FASEB J ; 34(4): 5262-5281, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32060981

RESUMEN

The neurotoxicity of hard metal-based nanoparticles (NPs) remains poorly understood. Here, we deployed the human neuroblastoma cell line SH-SY5Y differentiated or not into dopaminergic- and cholinergic-like neurons to study the impact of tungsten carbide (WC) NPs, WC NPs sintered with cobalt (Co), or Co NPs versus soluble CoCl2 . Co NPs and Co salt triggered a dose-dependent cytotoxicity with an increase in cytosolic calcium, lipid peroxidation, and depletion of glutathione (GSH). Co NPs and Co salt also suppressed glutathione peroxidase 4 (GPX4) mRNA and protein expression. Co-exposed cells were rescued by N-acetylcysteine (NAC), a precursor of GSH, and partially by liproxstatin-1, an inhibitor of lipid peroxidation. Furthermore, in silico analyses predicted a significant correlation, based on similarities in gene expression profiles, between Co-containing NPs and Parkinson's disease, and changes in the expression of selected genes were validated by RT-PCR. Finally, experiments using primary human dopaminergic neurons demonstrated cytotoxicity and GSH depletion in response to Co NPs and CoCl2 with loss of axonal integrity. Overall, these data point to a marked neurotoxic potential of Co-based but not WC NPs and show that neuronal cell death may occur through a ferroptosis-like mechanism.


Asunto(s)
Diferenciación Celular , Cobalto/química , Neuronas Dopaminérgicas/patología , Ferroptosis , Nanopartículas del Metal/toxicidad , Enfermedades Neurodegenerativas/patología , Células Cultivadas , Neuronas Dopaminérgicas/metabolismo , Glutatión/metabolismo , Humanos , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/química , Enfermedades Neurodegenerativas/inducido químicamente
9.
J Immunol ; 203(8): 2276-2290, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31519860

RESUMEN

Neutrophil extracellular traps (NETs) composed of nuclear DNA associated with histones and granule proteins are involved in the extracellular killing of pathogens. Excessive NET formation has been implicated in several noninfectious pathological conditions. The disposal of NETs is, therefore, important to prevent inadvertent effects resulting from the continued presence of NETs in the extracellular environment. In this study, we investigated the interaction of NETs released by freshly isolated, PMA-stimulated primary human neutrophils with primary human monocyte-derived macrophages or dendritic cells (DCs). NETs were internalized by macrophages, and removal of the protein component prevented engulfment of NETs, whereas complexation with LL-37 restored the uptake of "naked" (protein-free) NETs. NETs were also found to dampen the bacterial LPS-induced maturation of DCs. Cytokine profiling was conducted by using a multiplex array following the interaction of NETs with macrophages or DCs, and NETs alone were found to be noninflammatory, whereas immunomodulatory effects were noted in the presence of LPS with significant upregulation of IL-1ß secretion, and a marked suppression of other LPS-induced factors including vascular endothelial growth factor (VEGF) in both cell types. Moreover, macrophage digestion of NETs was dependent on TREX1 (also known as DNaseIII), but not DNaseII, whereas extracellular DNase1L3-mediated degradation of NETs was observed for DCs. Collectively, these findings shed light on the interactions between NETs and phagocytic cells and provide new insights regarding the clearance of NETs, double-edged swords of innate immunity.


Asunto(s)
Células Dendríticas/metabolismo , Trampas Extracelulares/metabolismo , Macrófagos/metabolismo , Neutrófilos/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Trampas Extracelulares/efectos de los fármacos , Voluntarios Sanos , Humanos , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Catelicidinas
10.
Carbon N Y ; 178: 563-572, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37206955

RESUMEN

Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) causes inflammation and fibrosis. Our previous work has shown that industrially produced MWCNTs trigger specific changes in gene expression in the lungs of exposed animals. To elucidate whether epigenetic effects play a role for these gene expression changes, we performed whole genome bisulphite sequencing to assess DNA methylation patterns in the lungs 56 days after exposure to MWCNTs. Lung tissues were also evaluated with respect to histopathological changes and cytokine profiling of bronchoalveolar lavage (BAL) fluid was conducted using a multi-plex array. Integrated analysis of transcriptomics data and DNA methylation data revealed concordant changes in gene expression. Functional analysis showed that the muscle contraction, immune system/inflammation, and extracellular matrix pathways were the most affected pathways. Taken together, the present study revealed that MWCNTs exert epigenetic effects in the lungs of exposed animals, potentially driving the subsequent gene expression changes.

11.
Semin Immunol ; 34: 33-51, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28869063

RESUMEN

The innate immune system provides the first line of defence against foreign microbes and particulate materials. Engineered nanoparticles can interact with the immune system in many different ways. Nanoparticles may thus elicit inflammation with engagement of neutrophils, macrophages and other effector cells; however, it is important to distinguish between acute and chronic inflammation in order to identify the potential hazards of nanoparticles for human health. Nanoparticles may also interact with and become internalised by dendritic cells, key antigen-presenting cells of the immune system, where a better understanding of these processes could pave the way for improved vaccination strategies. Nanoparticle characteristics such as size, shape and deformability also influence nanoparticle uptake by a plethora of immune cells and subsequent immune responses. Furthermore, the corona of adsorbed biomolecules on nanoparticle surfaces should not be neglected. Complement activation represents a special case of regulated and dynamic corona formation on nanoparticles with important implications in clearance and safety. Additionally, the inadvertent binding of bacterial lipopolysaccharide to nanoparticles is important to consider as this may skew the outcome and interpretation of immunotoxicological studies. Here, we discuss nanoparticle interactions with different cell types and soluble mediators belonging to the innate immune system.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Nanopartículas/metabolismo , Animales , Activación de Complemento , Humanos , Inflamación , Lipopolisacáridos/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Vacunación
12.
Acta Paediatr ; 110(11): 2912-2920, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34160857

RESUMEN

Congenital neutropenia with autosomal recessive inheritance was first described by the Swedish paediatrician Rolf Kostmann who coined the term 'infantile genetic agranulocytosis'. The condition is now commonly referred to as Kostmann disease. These patients display a maturation arrest of the myelopoiesis in the bone marrow and reduced neutrophil numbers and suffer from recurrent, often life-threatening infections. The molecular mechanism underlying congenital neutropenia has been intensively investigated, and mutations in genes that impinge on programmed cell death have been identified. The present review provides an overview of these studies.


Asunto(s)
Neutropenia , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Humanos , Mutación , Neutropenia/congénito , Neutropenia/genética , Síndrome
13.
Biochem Biophys Res Commun ; 533(1): 36-49, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-32921412

RESUMEN

Nanomedicine is at a crossroads: with relatively few success stories in terms of clinical translation despite more and more research on increasingly sophisticated nanomaterials, it is important to consider whether we are on the right track. Indeed, it is crucial that we address the fact that while considerable efforts are being made to overcome barriers to translation from the bench to the clinic, scientists are still struggling to decipher fundamental aspects of nanomaterial interactions with biological systems. We believe that a key to the successful adoption of nanomedicines in oncology and beyond lies in a deeper understanding of underlying biological processes and in decoding interactions between engineered nanomaterials and biological systems. Here we provide an overview of progress in nanomedicine during the past 5 years.


Asunto(s)
Nanomedicina/métodos , Nanoestructuras/uso terapéutico , Animales , Humanos , Nanopartículas de Magnetita/efectos adversos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Nanoestructuras/efectos adversos , Nanoestructuras/química , Nanotecnología/métodos , Neoplasias/terapia
14.
Small ; 16(21): e1907686, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32227449

RESUMEN

Numerous studies have addressed the biological impact of graphene-based materials including graphene oxide (GO), yet few have focused on long-term effects. Here, RNA sequencing is utilized to unearth responses of human lung cells to GO. To this end, the BEAS-2B cell line derived from normal human bronchial epithelium is subjected to repeated, low-dose exposures of GO (1 or 5 µg mL-1 ) for 28 days or to the equivalent, cumulative amount of GO for 48 h. Then, samples are analyzed by using the NovaSeq 6000 sequencing system followed by pathway analysis and gene ontology enrichment analysis of the differentially expressed genes. Significant differences are seen between the low-dose, long-term exposures and the high-dose, short-term exposures. Hence, exposure to GO for 48 h results in mitochondrial dysfunction. In contrast, exposure to GO for 28 days is characterized by engagement of apoptosis pathways with downregulation of genes belonging to the inhibitor of apoptosis protein (IAP) family. Validation experiments confirm that long-term exposure to GO affects the apoptosis threshold in lung cells, accompanied by a loss of IAPs. These studies reveal the sensitivity of RNA-sequencing approaches and show that acute exposure to GO is not a good predictor of the long-term effects of GO.


Asunto(s)
Exposición a Riesgos Ambientales , Grafito , Secuenciación de Nucleótidos de Alto Rendimiento , Pulmón , Apoptosis/efectos de los fármacos , Grafito/toxicidad , Humanos , Pulmón/efectos de los fármacos , Factores de Tiempo
15.
FASEB J ; 33(4): 4962-4974, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30657335

RESUMEN

Glucocorticoids (GCs) are frequently used to treat chronic disorders in children, including inflammation and cancer. Prolonged treatment with GCs is well known to impair bone growth, an effect linked to increased apoptosis and suppressed proliferation in growth plate chondrocytes. We hypothesized that the endogenous antiapoptotic protein humanin (HN) may prevent these effects. Interestingly, GC-induced bone growth impairment and chondrocyte apoptosis was prevented in HN overexpressing mice, HN-treated wild-type mice, and in HN-treated cultured rat metatarsal bones. GC-induced suppression of chondrocyte proliferation was also prevented by HN. Furthermore, GC treatment reduced Indian Hedgehog expression in growth plates of wild-type mice but not in HN overexpressing mice or HN-treated wild-type animals. A Hedgehog (Hh) antagonist, vismodegib, was found to suppress the growth of cultured rat metatarsal bones, and this effect was also prevented by HN. Importantly, HN did not interfere with the desired anti-inflammatory effects of GCs. We conclude that HN is a novel regulator of Hh signaling preventing GC-induced bone growth impairment without interfering with desired effects of GCs. Our data may open for clinical studies exploring a new possible strategy to prevent GC-induced bone growth impairment by cotreating with HN.-Zaman, F., Zhao, Y., Celvin, B., Mehta, H. H., Wan, J., Chrysis, D., Ohlsson, C., Fadeel, B., Cohen, P., Sävendahl, L. Humanin is a novel regulator of Hedgehog signaling and prevents glucocorticoid-induced bone growth impairment.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Glucocorticoides/farmacología , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Citocinas/metabolismo , Fragmentación del ADN/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Hedgehog/genética , Inmunohistoquímica , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Transducción de Señal/fisiología
16.
Nanomedicine ; 18: 112-121, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30849547

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related deaths and novel treatment approaches are urgently needed. Here we show that poly(ethylene glycol)-functionalized nanodiamonds loaded with doxorubicin (ND-PEG-DOX) afforded a considerable improvement over free drug in an orthotopic pancreatic xenograft model. ND-PEG-DOX complexes were also superior to free DOX in 3-dimensional (3D) tumor spheroids of PDAC. ND-PEG showed no cytotoxicity towards macrophages, and histopathological analysis showed no abnormalities of major organs upon in vivo administration of ND-PEG-DOX. These results provide evidence that ND-mediated drug delivery may serve as a means of improving the therapeutic outcome in PDAC.


Asunto(s)
Nanodiamantes/química , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Humanos , Hidrodinámica , Masculino , Ratones , Nanodiamantes/ultraestructura , Neoplasias Pancreáticas/ultraestructura , Tamaño de la Partícula , Polietilenglicoles/química , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Distribución Tisular/efectos de los fármacos , Resultado del Tratamiento
17.
Part Fibre Toxicol ; 15(1): 32, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30016969

RESUMEN

BACKGROUND: Genotoxicity is an important toxicological endpoint due to the link to diseases such as cancer. Therefore, an increased understanding regarding genotoxicity and underlying mechanisms is needed for assessing the risk with exposure to nanoparticles (NPs). The aim of this study was to perform an in-depth investigation regarding the genotoxicity of well-characterized Ni and NiO NPs in human bronchial epithelial BEAS-2B cells and to discern possible mechanisms. Comparisons were made with NiCl2 in order to elucidate effects of ionic Ni. METHODS: BEAS-2B cells were exposed to Ni and NiO NPs, as well as NiCl2, and uptake and cellular dose were investigated by transmission electron microscopy (TEM) and inductively coupled plasma mass spectrometry (ICP-MS). The NPs were characterized in terms of surface composition (X-ray photoelectron spectroscopy), agglomeration (photon cross correlation spectroscopy) and nickel release in cell medium (ICP-MS). Cell death (necrosis/apoptosis) was investigated by Annexin V-FITC/PI staining and genotoxicity by cytokinesis-block micronucleus (cytome) assay (OECD 487), chromosomal aberration (OECD 473) and comet assay. The involvement of intracellular reactive oxygen species (ROS) and calcium was explored using the fluorescent probes, DCFH-DA and Fluo-4. RESULTS: NPs were efficiently taken up by the BEAS-2B cells. In contrast, no or minor uptake was observed for ionic Ni from NiCl2. Despite differences in uptake, all exposures (NiO, Ni NPs and NiCl2) caused chromosomal damage. Furthermore, NiO NPs were most potent in causing DNA strand breaks and generating intracellular ROS. An increase in intracellular calcium was observed and modulation of intracellular calcium by using inhibitors and chelators clearly prevented the chromosomal damage. Chelation of iron also protected against induced damage, particularly for NiO and NiCl2. CONCLUSIONS: This study has revealed chromosomal damage by Ni and NiO NPs as well as Ni ionic species and provides novel evidence for a calcium-dependent mechanism of cyto- and genotoxicity.


Asunto(s)
Calcio/metabolismo , Aberraciones Cromosómicas/inducido químicamente , Pulmón/efectos de los fármacos , Mutágenos/toxicidad , Nanopartículas/toxicidad , Níquel/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular , Ensayo Cometa , Daño del ADN , Humanos , Pulmón/patología , Propiedades de Superficie
18.
J Appl Toxicol ; 38(3): 385-397, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29094763

RESUMEN

Our recent studies revealed a dose-dependent proinflammatory response to copper oxide nanoparticles (CuO NPs) in rats following short-term inhalation exposure for five consecutive days. Here transcriptomics approaches were applied using the same model to assess global gene expression in lung tissues obtained 1 day post-exposure and after a recovery period of 22 days from rats exposed to clean air or 6 hour equivalent doses of 3.3 mg m-3 (low dose) and 13.2 mg m-3 (high dose). Microarray analyses yielded about 1000 differentially expressed genes in the high-dose group and 200 in low-dose compared to the clean air control group, and less than 20 after the recovery period. Pathway analysis indicated cell proliferation/survival and inflammation as the main processes triggered by exposure to CuO NPs. We did not find significant perturbations of pathways related to oxidative stress. Upregulation of epithelial cell transforming protein 2 (Ect2), a known oncogene, was noted and ECT2 protein was upregulated in the lungs of exposed animals. Proliferation of alveolar epithelial cells was demonstrated based on Ki67 expression. The gene encoding monocyte chemoattractant protein 1 (or CCL2) was also upregulated and this was confirmed by immunohistochemistry. However, no aberrant DNA methylation of inflammation-associated genes was observed. In conclusion, we have found that inhalation of CuO NPs in rats causes upregulation of the oncoprotein ECT2 and the chemokine CCL2 and other proinflammatory markers as well as proliferation in bronchoalveolar epithelium after a short-term inhalation exposure. Thus, pathways known to be associated with neoplastic processes and inflammation were affected in this model.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cobre/toxicidad , Células Epiteliales/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Nanopartículas del Metal , Neumonía/inducido químicamente , Alveolos Pulmonares/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Administración por Inhalación , Animales , Proliferación Celular/genética , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Cobre/administración & dosificación , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Hiperplasia , Mediadores de Inflamación/metabolismo , Masculino , Neumonía/genética , Neumonía/metabolismo , Neumonía/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas Wistar , Factores de Tiempo
19.
Angew Chem Int Ed Engl ; 57(36): 11722-11727, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30006967

RESUMEN

Biodegradability of graphene is one of the fundamental parameters determining the fate of this material in vivo. Two types of aqueous dispersible graphene, corresponding to single-layer (SLG) and few-layer graphene (FLG), devoid of either chemical functionalization or stabilizing surfactants, were subjected to biodegradation by human myeloperoxidase (hMPO) mediated catalysis. Graphene biodegradation was also studied in the presence of activated, degranulating human neutrophils. The degradation of both FLG and SLG sheets was confirmed by Raman spectroscopy and electron microscopy analyses, leading to the conclusion that highly dispersed pristine graphene is not biopersistent.


Asunto(s)
Contaminantes Ambientales/metabolismo , Grafito/metabolismo , Peroxidasa/metabolismo , Biodegradación Ambiental , Contaminantes Ambientales/química , Proteínas Filagrina , Grafito/química , Humanos , Neutrófilos/enzimología , Neutrófilos/metabolismo , Espectrometría Raman
20.
J Proteome Res ; 16(2): 689-697, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27973853

RESUMEN

Thorough characterization of toxic effects of nanoparticles (NP) is desirable due to the increasing risk of potential environmental contamination by NP. In the current study, we combined three recently developed proteomics approaches to assess the effects of Au, CuO, and CdTe NP on the innate immune system. The human monocyte cell line THP-1 was employed as a model. The anticancer drugs camptothecin and doxorubicin were used as positive controls for cell death, and lipopolysaccharide was chosen as a positive control for proinflammatory activation. Despite equivalent overall toxicity effect (50 ± 10% dead cells), the three NP induced distinctly different proteomics signatures, with the strongest effect being induced by CdTe NP, followed by CuO and gold NP. The CdTe toxicity mechanism involves down-regulation of topoisomerases. The effect of CuO NP is most reminiscent of oxidative stress and involves up-regulation of proteins involved in heat response. The gold NP induced up-regulation of the inflammatory mediator, NF-κB, and its inhibitor TIPE2 was identified as a direct target of gold NP. Furthermore, gold NP triggered activation of NF-κB as evidenced by phosphorylation of the p65 subunit. Overall, the combined proteomics approach described here can be used to characterize the effects of NP on immune cells.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Inflamación/genética , Nanopartículas del Metal/efectos adversos , Proteoma/genética , Proteómica , Compuestos de Cadmio/efectos adversos , Camptotecina/administración & dosificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cobre/efectos adversos , Citotoxinas/efectos adversos , Doxorrubicina/administración & dosificación , Oro/efectos adversos , Humanos , Inmunidad Innata/genética , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteoma/efectos de los fármacos , Telurio/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA