Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
J Transl Med ; 22(1): 477, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764038

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a malignant tumor with a poor prognosis. Traditional treatments have limited effectiveness. Regulation of the immune response represents a promising new approach for OSCC treatment. B cells are among the most abundant immune cells in OSCC. However, the role of B cells in OSCC treatment has not been fully elucidated. METHODS: Single-cell RNA sequencing analysis of 13 tissues and 8 adjacent normal tissues from OSCC patients was performed to explore differences in B-cell gene expression between OSCC tissues and normal tissues. We further investigated the relationship between differentially expressed genes and the immune response to OSCC. We utilized tissue microarray data for 146 OSCC clinical samples and RNA sequencing data of 359 OSCC samples from The Cancer Genome Atlas (TCGA) to investigate the role of T-cell leukemia 1 A (TCL1A) in OSCC prognosis. Multiplex immunohistochemistry (mIHC) was employed to investigate the spatial distribution of TCL1A in OSCC tissues. We then investigated the effect of TCL1A on B-cell proliferation and trogocytosis. Finally, lentiviral transduction was performed to induce TCL1A overexpression in B lymphoblastoid cell lines (BLCLs) to verify the function of TCL1A. RESULTS: Our findings revealed that TCL1A was predominantly expressed in B cells and was associated with a better prognosis in OSCC patients. Additionally, we found that TCL1A-expressing B cells are located at the periphery of lymphatic follicles and are associated with tertiary lymphoid structures (TLS) formation in OSCC. Mechanistically, upregulation of TCL1A promoted the trogocytosis of B cells on dendritic cells by mediating the upregulation of CR2, thereby improving antigen-presenting ability. Moreover, the upregulation of TCL1A expression promoted the proliferation of B cells. CONCLUSION: This study revealed the role of B-cell TCL1A expression in TLS formation and its effect on OSCC prognosis. These findings highlight TCL1A as a novel target for OSCC immunotherapy.


Asunto(s)
Linfocitos B , Carcinoma de Células Escamosas , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca , Proteínas Proto-Oncogénicas , Estructuras Linfoides Terciarias , Humanos , Pronóstico , Neoplasias de la Boca/patología , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/inmunología , Estructuras Linfoides Terciarias/patología , Estructuras Linfoides Terciarias/inmunología , Estructuras Linfoides Terciarias/metabolismo , Linfocitos B/metabolismo , Linfocitos B/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Femenino , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Masculino , Persona de Mediana Edad , Línea Celular Tumoral , Proliferación Celular
2.
J Nanobiotechnology ; 22(1): 312, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840221

RESUMEN

Zinc oxide nanoparticles (ZNPs) are widely used in sunscreens and nanomedicines, and it was recently confirmed that ZNPs can penetrate stratum corneum into deep epidermis. Therefore, it is necessary to determine the impact of ZNPs on epidermis. In this study, ZNPs were applied to mouse skin at a relatively low concentration for one week. As a result, desmosomes in epidermal tissues were depolymerized, epidermal mechanical strain resistance was reduced, and the levels of desmosomal cadherins were decreased in cell membrane lysates and increased in cytoplasmic lysates. This finding suggested that ZNPs promote desmosomal cadherin endocytosis, which causes desmosome depolymerization. In further studies, ZNPs were proved to decrease mammalian target of rapamycin complex 1 (mTORC1) activity, activate transcription factor EB (TFEB), upregulate biogenesis of lysosome-related organelle complex 1 subunit 3 (BLOC1S3) and consequently promote desmosomal cadherin endocytosis. In addition, the key role of mTORC1 in ZNP-induced decrease in mechanical strain resistance was determined both in vitro and in vivo. It can be concluded that ZNPs reduce epidermal mechanical strain resistance by promoting desmosomal cadherin endocytosis via the mTORC1-TFEB-BLOC1S3 axis. This study helps elucidate the biological effects of ZNPs and suggests that ZNPs increase the risk of epidermal fragmentation.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Cadherinas , Endocitosis , Epidermis , Diana Mecanicista del Complejo 1 de la Rapamicina , Óxido de Zinc , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Endocitosis/efectos de los fármacos , Ratones , Cadherinas/metabolismo , Epidermis/metabolismo , Epidermis/efectos de los fármacos , Óxido de Zinc/farmacología , Óxido de Zinc/química , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Desmosomas/metabolismo , Nanopartículas/química , Estrés Mecánico
3.
J Nanobiotechnology ; 21(1): 140, 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37118804

RESUMEN

Atherosclerosis is one of the most common types of cardiovascular disease and is driven by lipid accumulation and chronic inflammation in the arteries, which leads to stenosis and thrombosis. Researchers have been working to design multifunctional nanomedicines with the ability to target, diagnose, and treat atherosclerosis, but recent studies have also identified that nanomaterials can cause atherosclerosis. Therefore, this review aims to outline the molecular mechanisms and physicochemical properties of nanomaterials that promote atherosclerosis. By analyzing the toxicological effects of nanomaterials on cells involved in the pathogenesis of atherosclerosis such as vascular endothelial cells, vascular smooth muscle cells and immune cells, we aim to provide new perspectives for the prevention and treatment of atherosclerosis, and raise awareness of nanotoxicology to advance the clinical translation and sustainable development of nanomaterials.


Asunto(s)
Aterosclerosis , Nanoestructuras , Humanos , Células Endoteliales , Aterosclerosis/inducido químicamente , Aterosclerosis/patología , Nanoestructuras/toxicidad , Nanoestructuras/química , Inflamación , Nanomedicina
4.
J Transl Med ; 20(1): 605, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36527141

RESUMEN

BACKGROUND: N6-methyladenosine (m6A) is the most prevalent epigenetic modification in eukaryotic messenger RNAs and plays a critical role in cell fate transition. However, it remains to be elucidated how m6A marks functionally impact the transcriptional cascades that orchestrate stem cell differentiation. The present study focuses on the biological function and mechanism of m6A methylation in dental pulp stem cell (DPSC) differentiation. METHODS: m6A RNA immunoprecipitation sequencing was utilized to assess the m6A-mRNA landscape during DPSC differentiation. Ectopic transplantation of DPSCs in immunodeficient mice was conducted to verify the in vitro findings. RNA sequencing and m6A RNA immunoprecipitation sequencing were combined to identify the candidate targets. RNA immunoprecipitation and RNA/protein stability of Noggin (NOG) were evaluated. The alteration in poly(A) tail was measured by 3'-RACE and poly(A) tail length assays. RESULTS: We characterized a dynamic m6A-mRNA landscape during DPSC mineralization with increasing enrichment in the 3' untranslated region (UTR). Methyltransferase-like 3 (METTL3) was identified as the key m6A player, and METTL3 knockdown disrupted functional DPSC differentiation. Moreover, METTL3 overexpression enhanced DPSC mineralization. Increasing m6A deposition in the 3' UTR restricted NOG expression, which is required for DPSC mineralization. This stage-specific m6A methylation and destabilization of NOG was suppressed by METTL3 knockdown only in differentiated DPSCs. Furthermore, METTL3 promotes the degradation of m6A-tagged NOG by shortening the poly(A) tail length in the differentiated stage. CONCLUSIONS: Our results address an essential role of dynamic m6A signaling in the temporal control of DPSC differentiation and provide new insight into epitranscriptomic mechanisms in stem cell-based therapy.


Asunto(s)
Adenosina , Metiltransferasas , Ratones , Animales , Metiltransferasas/genética , Metiltransferasas/metabolismo , Adenosina/metabolismo , Pulpa Dental , Diferenciación Celular , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
J Nanobiotechnology ; 20(1): 265, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672765

RESUMEN

Overproduced reactive oxygen and reactive nitrogen species (RONS) in the brain are involved in the pathogenesis of several neurological diseases, such as Alzheimer's disease, Parkinson's disease, traumatic brain injury, and stroke, as they attack neurons and glial cells, triggering cellular redox stress. Neutralizing RONS, and, thus, alleviating redox stress, can slow down or stop the progression of neurological diseases. Currently, an increasing number of studies are applying nanomaterials (NMs) with anti-redox activity and exploring the potential mechanisms involved in redox stress-related neurological diseases. In this review, we summarize the anti-redox mechanisms of NMs, including mimicking natural oxidoreductase activity and inhibiting RONS generation at the source. In addition, we propose several strategies to enhance the anti-redox ability of NMs and highlight the challenges that need to be resolved in their application. In-depth knowledge of the mechanisms and potential application of NMs in alleviating redox stress will help in the exploration of the therapeutic potential of anti-redox stress NMs in neurological diseases.


Asunto(s)
Nanoestructuras , Especies de Nitrógeno Reactivo , Antioxidantes/uso terapéutico , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno
6.
J Nanobiotechnology ; 19(1): 108, 2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863340

RESUMEN

The widespread use of nanomaterials (NMs) has raised concerns that exposure to them may introduce potential risks to the human body and environment. The liver is the main target organ for NMs. Hepatotoxic effects caused by NMs have been observed in recent studies but have not been linked to liver disease, and the intrinsic mechanisms are poorly elucidated. Additionally, NMs exhibit varied toxicokinetics and induce enhanced toxic effects in susceptible livers; however, thus far, this issue has not been thoroughly reviewed. This review provides an overview of the toxicokinetics of NMs. We highlight the possibility that NMs induce hepatic diseases, including nonalcoholic steatohepatitis (NASH), fibrosis, liver cancer, and metabolic disorders, and explore the underlying intrinsic mechanisms. Additionally, NM toxicokinetics and the potential induced risks in the livers of susceptible individuals, including subjects with liver disease, obese individuals, aging individuals and individuals of both sexes, are summarized. To understand how NM type affect their toxicity, the influences of the physicochemical and morphological (PCM) properties of NMs on their toxicokinetics and toxicity are also explored. This review provides guidance for further toxicological studies on NMs and will be important for the further development of NMs for applications in various fields.


Asunto(s)
Hepatopatías/metabolismo , Hígado/metabolismo , Nanoestructuras/química , Nanoestructuras/toxicidad , Animales , Fibrosis , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Neoplasias Hepáticas , Enfermedades Metabólicas , Toxicocinética
7.
Part Fibre Toxicol ; 17(1): 53, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33066795

RESUMEN

BACKGROUND: Widespread biomedical applications of nanomaterials (NMs) bring about increased human exposure risk due to their unique physicochemical properties. Autophagy, which is of great importance for regulating the physiological or pathological activities of the body, has been reported to play a key role in NM-driven biological effects both in vivo and in vitro. The coexisting hazard and health benefits of NM-mediated autophagy in biomedicine are nonnegligible and require our particular concerns. MAIN BODY: We collected research on the toxic effects related to NM-mediated autophagy both in vivo and in vitro. Generally, NMs can be delivered into animal models through different administration routes, or internalized by cells through different uptake pathways, exerting varying degrees of damage in tissues, organs, cells, and organelles, eventually being deposited in or excreted from the body. In addition, other biological effects of NMs, such as oxidative stress, inflammation, necroptosis, pyroptosis, and ferroptosis, have been associated with autophagy and cooperate to regulate body activities. We therefore highlight that NM-mediated autophagy serves as a double-edged sword, which could be utilized in the treatment of certain diseases related to autophagy dysfunction, such as cancer, neurodegenerative disease, and cardiovascular disease. Challenges and suggestions for further investigations of NM-mediated autophagy are proposed with the purpose to improve their biosafety evaluation and facilitate their wide application. Databases such as PubMed and Web of Science were utilized to search for relevant literature, which included all published, Epub ahead of print, in-process, and non-indexed citations. CONCLUSION: In this review, we focus on the dual effect of NM-mediated autophagy in the biomedical field. It has become a trend to use the benefits of NM-mediated autophagy to treat clinical diseases such as cancer and neurodegenerative diseases. Understanding the regulatory mechanism of NM-mediated autophagy in biomedicine is also helpful for reducing the toxic effects of NMs as much as possible.


Asunto(s)
Nanoestructuras/toxicidad , Animales , Autofagia/fisiología , Tecnología Biomédica , Humanos , Neoplasias , Enfermedades Neurodegenerativas , Estrés Oxidativo
8.
J Nanobiotechnology ; 18(1): 52, 2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32188458

RESUMEN

BACKGROUND: Graphene oxide (GO) nanoparticles (NPs) have been widely applied in various fields, especially in biomedical applications. Extensive studies have suggested that GO can pass through the blood-brain barrier (BBB) and induce abnormal autophagy and cytotoxicity in the central nervous system (CNS). However, the effect and specific mechanism of GO on astrocytes, the most abundant cells in the brain still has not been extensively investigated. RESULTS: In this study, we systematically explored the toxicity and mechanism of GO exposure in the rat astroglioma-derived F98 cell line using molecular biological techniques (immunofluorescence staining, flow cytometry and Western blot) at the subcellular level and the signaling pathway level. Cells exposed to GO exhibited decreased cell viability and increased lactate dehydrogenase (LDH) release in a concentration- and time-dependent manner. GO-induced autophagy was evidenced by transmission electron microscopy (TEM) and immunofluorescence staining. Western blots showed that LC3II/I and p62 were upregulated and PI3K/Akt/mTOR was downregulated. Detection of lysosomal acidity and cathepsin B activity assay indicated the impairment of lysosomal function. Annexin V-FITC-PI detection showed the occurrence of apoptosis after GO exposure. The decrease in mitochondrial membrane potential (MMP) with an accompanying upregulation of cleaved caspase-3 and Bax/Bcl-2 further suggested that endogenous signaling pathways were involved in GO-induced apoptosis. CONCLUSION: The exposure of F98 cells to GO can elicit concentration- and time-dependent toxicological effects. Additionally, increased autophagic response can be triggered after GO treatment and that the blocking of autophagy flux plays a vital role in GO cytotoxicity, which was determined to be related to dysfunction of lysosomal degradation. Importantly, the abnormal accumulation of autophagic substrate p62 protein can induce capase-3-mediated apoptosis. Inhibition of abnormal accumulation of autophagic cargo could alleviate the occurrence of GO-induced apoptosis in F98 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Grafito/farmacología , Lisosomas/efectos de los fármacos , Nanopartículas/química , Animales , Anexinas , Astrocitos , Caspasa 3/metabolismo , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
9.
J Nanobiotechnology ; 18(1): 9, 2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31918719

RESUMEN

The vascular system, which transports oxygen and nutrients, plays an important role in wound healing, cardiovascular disease treatment and bone tissue engineering. Angiogenesis is a complex and delicate regulatory process. Vascular cells, the extracellular matrix (ECM) and angiogenic factors are indispensable in the promotion of lumen formation and vascular maturation to support blood flow. However, the addition of growth factors or proteins involved in proangiogenic effects is not effective for regulating angiogenesis in different microenvironments. The construction of biomaterial scaffolds to achieve optimal growth conditions and earlier vascularization is undoubtedly one of the most important considerations and major challenges among engineering strategies. Nanomaterials have attracted much attention in biomedical applications due to their structure and unique photoelectric and catalytic properties. Nanomaterials not only serve as carriers that effectively deliver factors such as angiogenesis-related proteins and mRNA but also simulate the nano-topological structure of the primary ECM of blood vessels and stimulate the gene expression of angiogenic effects facilitating angiogenesis. Therefore, the introduction of nanomaterials to promote angiogenesis is a great helpful to the success of tissue regeneration and some ischaemic diseases. This review focuses on the angiogenic effects of nanoscaffolds in different types of tissue regeneration and discusses the influencing factors as well as possible related mechanisms of nanomaterials in endothelial neovascularization. It contributes novel insights into the design and development of novel nanomaterials for vascularization and therapeutic applications.


Asunto(s)
Nanoestructuras/química , Neovascularización Fisiológica , Animales , Autofagia , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Regeneración , Transducción de Señal
10.
Arch Toxicol ; 94(5): 1479-1495, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32180035

RESUMEN

Nanoparticles induce neurotoxicity following inhalation, oral administration, intravenous administration, or injection. Different pathways have various corresponding characteristics. Among them, the sensory nerve-to-brain pathways, which are direct neural pathways, bypass barriers such as the blood-brain barrier, which prevents the entry of the majority of nanoparticles into the brain. Subsequently, nanoparticles exert effects on sensory neuroreceptors and sensory nerves, causing central neurotoxicity. However, no studies have summarized sensory nerve-to-brain pathways for transporting nanoparticles. Here, we review recent findings on the potential sensory nerve pathways that promote nanoparticle entry into the brain, the effects of NPs on sensory receptors and sensory nerves, the central neurotoxicity induced by nanoparticles via sensory nerve pathways, and the possible mechanisms underlying these effects. In addition, the limitations of current research and possible trends for future research are also discussed. In summary, we hope that this review will serve as a reference, inspire ideas for further research into the neurotoxicity of nanoparticles, and facilitate the development of protective measures and treatment schemes for nanoparticle-induced neurotoxicity.


Asunto(s)
Encéfalo/fisiología , Nanopartículas/toxicidad , Sistema Nervioso/efectos de los fármacos , Animales , Barrera Hematoencefálica , Humanos , Síndromes de Neurotoxicidad
11.
Small ; 15(25): e1901073, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31062916

RESUMEN

Autophagy is a biological process that has attracted considerable attention as a target for novel therapeutics. Recently, nanomaterials (NMs) have been reported to modulate autophagy, which makes them potential agents for the treatment of autophagy-related diseases. In this study, zinc oxide nanoparticles (ZNPs) are utilized to evaluate NM-induced autophagy and debate the mechanisms involved. It is found that ZNPs undergo pH-dependent ion shedding and that intracellular zinc ions (Zn2+ ) play a crucial role in autophagy. Autophagy is activated with ZNPs treatment, which is inhibited after Zn2+ sequestration via ethylenediamine tetra-acetic acid. Lysosome-based autophagic degradation is halted after ZNPs treatment for more than 3 h and is accompanied by blockage of lysophagy, which renews impaired lysosomes. Furthermore, the microtubule (MT) system participates in ZNP-induced lysosome-autophagy system changes, especially in the fusion between autophagosomes and lysosomes. MT acetylation is helpful for protecting from ZNP-induced MT disruption, and it promotes the autophagic degradation process. In conclusion, this study provides valuable information on NM-induced lysosome-autophagy system changes, particularly with respect to the role of lysophagy and the MT system, which point to some attractive targets for the design of engineered nanoparticles.


Asunto(s)
Autofagia , Lisosomas/metabolismo , Microtúbulos/metabolismo , Nanopartículas/química , Óxido de Zinc/química , Acetilación , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Iones , Lisosomas/ultraestructura , Microtúbulos/ultraestructura , Nanopartículas/ultraestructura , Células PC12 , Ratas , Zinc/metabolismo
12.
Acta Biochim Biophys Sin (Shanghai) ; 51(12): 1208-1215, 2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31735958

RESUMEN

Emerging evidence shows that the long noncoding RNA taurine-upregulated gene 1 (TUG1) plays pivotal roles in regulating biological properties and functions of parenchyma cells in various types of disease processes. However, the mechanism underlying the effects of TUG1 on cell proliferation and apoptosis of human periodontal ligament cells (PDLCs) in periodontitis is undefined. In this study, we explored the functions of TUG1 and its underlying mechanisms in the inflammatory process induced by Porphyromonas gingivalis-derived lipopolysaccharide (LPS) in PDLCs. Our results showed that TUG1 had a decreased expression in both periodontal ligament (PDL) tissues with periodontitis and PDLCs under a LPS-induced inflammatory condition, and TUG1 expression was negatively correlated with miR-132 expression in periodontitis-affected PDL tissues. Furthermore, we found that TUG1 overexpression in PDLCs alleviated LPS-induced proliferative inhibition and apoptosis promotion, while TUG1 knockdown had the opposite effect. In addition, miR-132 inhibitor alleviated TUG1 knockdown-induced inhibition of proliferation and increase of apoptosis in PDLCs under inflammatory conditions induced by LPS. These findings indicated that TUG1 has an enormous potential in regulating cell proliferation and apoptosis of PDLCs during periodontitis and may provide an effective therapeutic target for periodontitis to reduce the damage caused by inflammatory reactions.


Asunto(s)
Infecciones por Bacteroidaceae/metabolismo , MicroARNs/metabolismo , Ligamento Periodontal , Periodontitis/metabolismo , ARN Largo no Codificante/fisiología , Adulto , Apoptosis , Proliferación Celular , Voluntarios Sanos , Humanos , Lipopolisacáridos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/patología , Porphyromonas gingivalis/patogenicidad , Adulto Joven
13.
Part Fibre Toxicol ; 15(1): 39, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340606

RESUMEN

BACKGROUND: The extensive biological applications of zinc oxide nanoparticles (ZnO NPs) in stomatology have created serious concerns about their biotoxicity. In our previous study, ZnO NPs were confirmed to transfer to the central nervous system (CNS) via the taste nerve pathway and cause neurodegeneration after 30 days of tongue instillation. However, the potential adverse effects on the brain caused by tongue-instilled ZnO NPs are not fully known. METHODS: In this study, the biodistribution of Zn, cerebral histopathology and inflammatory responses were analysed after 30 days of ZnO NPs tongue instillation. Moreover, the molecular mechanisms underlying neuroinflammation in vivo were further elucidated by treating BV2 and PC12 cells with ZnO NPs in vitro. RESULTS: This analysis indicated that ZnO NPs can transfer into the CNS, activate glial cells and cause neuroinflammation after tongue instillation. Furthermore, exposure to ZnO NPs led to a reduction in cell viability and induction of inflammatory response and calcium influx in BV2 and PC12 cells. The mechanism underlying how ZnO NPs induce neuroinflammation via the Ca2+-dependent NF-κB, ERK and p38 activation pathways was verified at the cytological level. CONCLUSION: This study provided a new way how NPs, such as ZnO NPs, induce neuroinflammation via the taste nerve translocation pathway, a new mechanism for ZnO NPs-induced neuroinflammation and a new direction for nanomaterial toxicity analysis.


Asunto(s)
Encéfalo/efectos de los fármacos , Calcio/metabolismo , Nanopartículas/toxicidad , Síndromes de Neurotoxicidad/inmunología , Lengua/efectos de los fármacos , Óxido de Zinc/toxicidad , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/genética , Expresión Génica/efectos de los fármacos , Inflamación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , FN-kappa B/metabolismo , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Células PC12 , Ratas , Ratas Wistar , Distribución Tisular , Lengua/inmunología , Lengua/metabolismo , Óxido de Zinc/farmacocinética
14.
Cell Physiol Biochem ; 40(6): 1487-1505, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997890

RESUMEN

Inorganic nanomaterials have been widely applied in biomedicine. However, several studies have noted that inorganic nanoparticles can enter the brain and induce cytoskeletal remodeling, as well as electrophysiological alterations, which are related to neurodevelopmental disorders and neurodegenerative diseases. The toxic effects of inorganic nanomaterials on the cytoskeleton and electrophysiology are summarized in this review. The relationships between inorganic NPs-induced cytoskeletal and electrophysiological alterations in the central nervous system remain obscure. We propose several potential relationships, including those involving N-methyl-D-aspartate receptor function, ion channels, transient receptor potential channels, and the Rho pathway.


Asunto(s)
Sistema Nervioso Central/fisiopatología , Citoesqueleto/metabolismo , Fenómenos Electrofisiológicos , Compuestos Inorgánicos/toxicidad , Nanopartículas/toxicidad , Animales , Sistema Nervioso Central/efectos de los fármacos , Humanos , Neurotransmisores/metabolismo
15.
Crit Rev Toxicol ; 46(4): 348-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963861

RESUMEN

Zinc oxide nanoparticles (ZnO NPs) are nanomaterials that are widely used in many fields. ZnO NPs are ion-shedding particles, and zinc ions produce important and potent effects that differ from those of other metal or metal oxide NPs. Several studies have reported the toxicological effects of ZnO NPs administered via several different routes, including orally, dermally, by pulmonary absorption, intraperitoneally, and intravenously. Some potential routes for human exposure have produced various toxic effects in animal models. Moreover, several in vitro studies using a range of cell lines have reported the mechanisms underlying ZnO NP toxicity. Zinc ions play a very important role in ZnO NP toxicity, although the effects of the particulate form cannot be excluded. A crucial determinant of toxicity is the solubility of ZnO NPs, which is influenced by various factors, including the pH of the environment in tissues, cells, and organelles. In addition to the inflammatory responses and oxidative stress known to be induced by ZnO NPs, these NPs also exhibit some positive anti-inflammatory, anti-diabetic, and pro-coagulant effects at sub-toxic doses; these effects are probably induced by zinc ions, which are an essential element in cell homeostasis. It is highly likely that there are additional distinct mechanisms at sub-toxic doses and concentrations, which may be concealed or altered by the toxic effects observed at higher levels of ZnO NPs. Furthermore, many signaling pathway molecules associated with necrosis and apoptosis can be activated, leading to cell death. This review presents the status of ZnO NP toxicology and highlights areas requiring further investigation.


Asunto(s)
Nanopartículas/toxicidad , Óxido de Zinc/toxicidad , Animales , Humanos , Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/envenenamiento , Óxido de Zinc/química , Óxido de Zinc/farmacocinética , Óxido de Zinc/envenenamiento
16.
Part Fibre Toxicol ; 13(1): 57, 2016 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-27799056

RESUMEN

Due to their unique physicochemical properties, graphene-family nanomaterials (GFNs) are widely used in many fields, especially in biomedical applications. Currently, many studies have investigated the biocompatibility and toxicity of GFNs in vivo and in intro. Generally, GFNs may exert different degrees of toxicity in animals or cell models by following with different administration routes and penetrating through physiological barriers, subsequently being distributed in tissues or located in cells, eventually being excreted out of the bodies. This review collects studies on the toxic effects of GFNs in several organs and cell models. We also point out that various factors determine the toxicity of GFNs including the lateral size, surface structure, functionalization, charge, impurities, aggregations, and corona effect ect. In addition, several typical mechanisms underlying GFN toxicity have been revealed, for instance, physical destruction, oxidative stress, DNA damage, inflammatory response, apoptosis, autophagy, and necrosis. In these mechanisms, (toll-like receptors-) TLR-, transforming growth factor ß- (TGF-ß-) and tumor necrosis factor-alpha (TNF-α) dependent-pathways are involved in the signalling pathway network, and oxidative stress plays a crucial role in these pathways. In this review, we summarize the available information on regulating factors and the mechanisms of GFNs toxicity, and propose some challenges and suggestions for further investigations of GFNs, with the aim of completing the toxicology mechanisms, and providing suggestions to improve the biological safety of GFNs and facilitate their wide application.


Asunto(s)
Grafito/toxicidad , Nanopartículas/toxicidad , Animales , Vías de Administración de Medicamentos , Distribución Tisular
17.
J Mater Sci Mater Med ; 27(3): 59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26800690

RESUMEN

Inflammatory effects are significant elements of the immune response to biomaterials. Previously, we reported inflammatory effects in response to dicalcium silicate (Ca2SiO4, C2S) particles. However, the immunological effects of C2S coatings have not been studied. C2S often used as coatings materials in orthopedic and dentistry applications. It may have different effect from C2S particles. Further, it remains unclear whether C2S coating is equally biocompatible as 45S5 coating. The aim of this study was to test the cytotoxicity and pro-inflammatory effects of C2S coating on RAW 264.7 macrophages. C2S and 45S5 coatings were characterized using scanning electron microscopy (SEM), atomic force microscopy (AFM), energy dispersive analysis (EDS) and X-ray diffraction (XRD). inductively coupled plasma optical emission spectroscopy (ICP-OES) was used to detect ionic concentrations after soaking coated discs in medium. The cytotoxicity of C2S and 45S5 coatings against RAW 264.7 macrophages was measured using the LDH Cytotoxicity Assay Kit, Cell Counting Kit-8 (CCK-8) assays and flow cytometry for apoptosis assays. The gene and protein expression of TNF-α, IL-6 and IL-1ß were detected using RT-q PCR and ELISA, respectively. The tested coating materials are not cytotoxic to macrophages. The C2S-coated surface stimulated macrophages to express pro-inflammatory mediators, such as TNF-α, IL-6 and IL-1ß, and C2S coating caused less IL-6 but greater IL-1ß production than the 45S5 coating. C2S coating have no cytotoxicity when directly cultured with macrophages. C2S and 45S5 coatings both have the potential to induce pro-inflammatory effects, and the biocompatibility of C2S is similar to that of 45S5.


Asunto(s)
Compuestos de Calcio/toxicidad , Supervivencia Celular/efectos de los fármacos , Inflamación/inducido químicamente , Macrófagos/efectos de los fármacos , Silicatos/toxicidad , Animales , Compuestos de Calcio/química , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Ensayo de Materiales , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Silicatos/química , Andamios del Tejido
19.
J Prosthet Dent ; 112(6): 1406-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24993379

RESUMEN

STATEMENT OF PROBLEM: Zirconia posts exhibit high strength and toughness, but reliable bonding with the resin core is difficult to attain. The use of a ferrule has been found to improve stress distribution in the root of endodontically treated teeth. PURPOSE: The purpose of this finite element analysis study was to measure the stress distribution in the post-core system and root structure of a maxillary canine. MATERIAL AND METHODS: A right maxillary canine was embedded and subjected to a micro-computed tomography scan. Three-dimensional dynamic scan data were then transformed, and a finite element model of 4 dentin ferrule heights was designed with zirconia posts and heat-pressed glass ceramics for a complete crown restoration. Von Mises stresses were determined by applying a 300 N static load to the middle of the lingual surface of the crown. RESULTS: When the ferrule height increased from 0 to 3 mm, the maximum von Mises stress of the zirconia post decreased from 196 to 149 MPa, and that on the zirconia post-dentin interface decreased from 174 to 132 MPa. The maximum von Mises stress decreased from 39.8 to 32.5 MPa in the apical root and from 59.5 to 49.9 MPa in the mid-root when the ferrule height increased from 0 to 3 mm. CONCLUSIONS: Increased ferrule height is associated with reduced von Mises stress in the zirconia post and the post-dentin interface, with an apparent shift of von Mises stress to the root cervical area from the mid-root and the apex.


Asunto(s)
Diente Canino/patología , Materiales Dentales/química , Análisis de Elementos Finitos , Imagenología Tridimensional/métodos , Técnica de Perno Muñón/instrumentación , Circonio/química , Cadáver , Cerámica/química , Simulación por Computador , Coronas , Diseño de Prótesis Dental , Dentina/patología , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Ensayo de Materiales , Maxilar , Modelos Biológicos , Estrés Mecánico , Ápice del Diente/patología , Cuello del Diente/patología , Raíz del Diente/patología , Microtomografía por Rayos X/métodos
20.
Toxicology ; 504: 153803, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38616010

RESUMEN

Nanomaterials are widely utilized in several domains, such as everyday life, societal manufacturing, and biomedical applications, which expand the potential for nanomaterials to penetrate biological barriers and interact with cells. Multiple studies have concentrated on the particular or improper utilization of nanomaterials, resulting in cellular death. The primary mode of cell death caused by nanotoxicity is programmable cell death, which includes apoptosis, ferroptosis, necroptosis, and pyroptosis. Based on our prior publications and latest research, mitochondria have a vital function in facilitating programmed cell death caused by nanomaterials, as well as initiating or transmitting death signal pathways associated with it. Therefore, this review takes mitochondria as the focal point to investigate the internal molecular mechanism of nanomaterial-induced programmed cell death, with the aim of identifying potential targets for prevention and treatment in related studies.


Asunto(s)
Apoptosis , Mitocondrias , Nanoestructuras , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanoestructuras/toxicidad , Animales , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda