Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biometals ; 37(3): 609-629, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38277035

RESUMEN

Cadmium (Cd) is an important environmental pollutant that poses a threat to human health and represents a critical component of air pollutants, food sources, and cigarette smoke. Cd is a known carcinogen and has toxic effects on the environment and various organs in humans. Heavy metals within an organism are difficult to biodegrade, and those that enter the respiratory tract are difficult to remove. Autophagy is a key mechanism for counteracting extracellular (microorganisms and foreign bodies) or intracellular (damaged organelles and proteins that cannot be degraded by the proteasome) stress and represents a self-protective mechanism for eukaryotes against heavy metal toxicity. Autophagy maintains cellular homeostasis by isolating and gathering information about foreign chemicals associated with other molecular events. However, autophagy may trigger cell death under certain pathological conditions, including cancer. Autophagy dysfunction is one of the main mechanisms underlying Cd-induced cytotoxicity. In this review, the toxic effects of Cd-induced autophagy on different human organ systems were evaluated, with a focus on hepatotoxicity, nephrotoxicity, respiratory toxicity, and neurotoxicity. This review also highlighted the classical molecular pathways of Cd-induced autophagy, including the ROS-dependent signaling pathways, endoplasmic reticulum (ER) stress pathway, Mammalian target of rapamycin (mTOR) pathway, Beclin-1 and Bcl-2 family, and recently identified molecules associated with Cd. Moreover, research directions for Cd toxicity regarding autophagic function were proposed. This review presents the latest theories to comprehensively reveal autophagy behavior in response to Cd toxicity and proposes novel potential autophagy-targeted prevention and treatment strategies for Cd toxicity and Cd-associated diseases in humans.


Asunto(s)
Autofagia , Cadmio , Autofagia/efectos de los fármacos , Humanos , Cadmio/toxicidad , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Contaminantes Ambientales/toxicidad
2.
Environ Res ; 242: 117733, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38000634

RESUMEN

Carbon black nanoparticles (CBNPs) and cadmium (Cd) are major components of various air pollutants and cigarette smoke. Autophagy and inflammation both play critical roles in understanding the toxicity of particles and their components, as well as maintaining body homeostasis. However, the effects and mechanisms of CBNPs and Cd (CBNPs-Cd) co-exposure on the human respiratory system remain unclear. In this study, a CBNPs-Cd exposure model was constructed to explore the respiratory toxicity and combined mechanism of these chemicals on the autophagy-lysosome pathway in the context of respiratory inflammation. Co-exposure of CBNPs and Cd significantly increased the number of autophagosomes and lysosomes in human bronchial epithelial cells (16HBE) and mouse lung tissues compared to the control group, as well as the groups exposed to CBNPs and Cd alone. Autophagic markers, LC3II and P62 proteins, were up-regulated in 16HBE cells and mouse lung tissues after CBNPs-Cd co-exposure. However, treatment with Cq inhibitor (an indicator of lysosomal acid environment) resulted in a substantial decreased co-localization fluorescence of LC3 and lysosomes in the CBNPs-Cd combination group compared with the CBNPs-Cd single and control groups. No difference in LAMP1 protein expression was observed among the exposed groups. Adding 3 MA alleviated inflammatory responses, while applying the Baf-A1 inhibitor aggravated inflammation both in vitro and in vivo following CBNPs-Cd co-exposure. Factorial analysis showed no interaction between CBNPs and Cd in their effects on 16HBE cells. We demonstrated that co-exposure to CBNPs-Cd increases the synthesis of autophagosomes and regulates the acidic environment of lysosomes, thereby inhibiting autophagy-lysosome fusion and enhancing the inflammatory response in both 16HBE cells and mouse lung. These findings provide evidence for a comprehensive understanding of the interaction between CBNPs and Cd in mixed pollutants, as well as for the prevention and control of occupational exposure to these two chemicals.


Asunto(s)
Cadmio , Nanopartículas , Ratones , Humanos , Animales , Cadmio/toxicidad , Hollín/toxicidad , Autofagia , Inflamación/inducido químicamente , Inflamación/metabolismo , Células Epiteliales , Lisosomas/metabolismo , Nanopartículas/toxicidad
3.
Environ Toxicol ; 39(1): 377-387, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37782690

RESUMEN

Fine particulate matter (PM2.5 ) has been shown to induce lung injury. However, the pathophysiological mechanisms of PM2.5 -induced pulmonary injury after different exposure times are poorly understood. In this study, we exposed male ICR mice to a whole-body PM2.5 inhalation system at daily mean concentration range from 92.00 to 862.00 µg/m3 for 30, 60, and 90 days. We found that following prolonged exposure to PM2.5 , pulmonary injury was increasingly evident with significant histopathological alterations. Notably, the pulmonary inflammatory response and fibrosis caused by PM2.5 after different exposure times were closely associated with histopathological changes. In addition, PM2.5 exposure caused oxidative stress, DNA damage and impairment of DNA repair in a time-dependent manner in the lung. Importantly, exposure to PM2.5 eventually caused apoptosis in the lung through upregulation of cleaved-caspase-3 and downregulation of Bcl-2. Overall, our data demonstrated that PM2.5 led to pulmonary injury in a time-dependent manner via upregulation of proinflammatory and fibrosis-related genes, and activation of the DNA damage response. Our findings provided a novel perspective on the pathophysiology of respiratory diseases caused by airborne pollution.


Asunto(s)
Lesión Pulmonar , Ratones , Masculino , Animales , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones Endogámicos ICR , Material Particulado/toxicidad , Pulmón/patología , Estrés Oxidativo/genética , Fibrosis
4.
Gerontology ; 69(9): 1065-1075, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37285833

RESUMEN

INTRODUCTION: Alzheimer's disease (AD) is a major public health concern worldwide, but there are still no drugs available that treat it effectively. Previous studies have shown that phenylethanoid glycosides have pharmacological effects, which include anti-AD properties, but the underlying mechanisms by which they ameliorate AD symptoms remain unknown. METHODS: In this study, we used an APP/PS1 AD mouse model to explore the function and mechanisms underlying savatiside A (SA) and torenoside B (TB) in the treatment of AD. SA or TB (100 mg·kg-1·d-1) was orally administered to 7-month-old APP/PS1 mice for 4 weeks. Cognitive and memory functions were measured using behavioral experiments (including the Morris water maze test and the Y-maze spontaneous alternation test). Molecular biology experiments (including Western blotting, immunofluorescence, and enzyme-linked immunosorbent assays) were used to detect any corresponding changes in signaling pathways. RESULTS: The results showed that SA or TB treatment could significantly reduce cognitive impairment in APP/PS1 mice. We also showed that chronic treatment with SA/TB could prevent spine loss, synaptophysin immunoreactivity, and neuronal loss in mice, thereby improving synaptic plasticity and moderating learning and memory deficits. SA/TB administration also promoted the expression of synaptic proteins in APP/PS1 mouse brains and upregulated phosphorylation of proteins in the cyclic adenosine monophosphate (cAMP)/CREB/brain-derived neurotrophic growth factor (BDNF) pathway that are responsible for synaptic plasticity. Additionally, chronic SA/TB treatment increased the levels of BDNF and nerve growth factor (NGF) in the brains of APP/PS1 mice. Both astrocyte and microglia volumes, as well as the generation of amyloid ß, were also decreased in SA/TB-treated APP/PS1 mice compared to control APP/PS1 mice. CONCLUSION: In summary, SA/TB treatment was associated with activation of the cAMP/CREB/BDNF pathway and increased BDNF and NGF expression, indicating that SA/TB improves cognitive functioning via nerve regeneration. SA/TB is a promising candidate drug for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratones , Animales , Ratones Transgénicos , Péptidos beta-Amiloides/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/farmacología , Factor de Crecimiento Nervioso/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal , Encéfalo/metabolismo , Aprendizaje por Laberinto , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Modelos Animales de Enfermedad
5.
J Environ Sci (China) ; 124: 451-461, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36182153

RESUMEN

Inflammation is a major adverse outcome induced by inhaled particulate matter with a diameter of ≤ 2.5 µm (PM2.5), and a critical trigger of most PM2.5 exposure-associated diseases. However, the key molecular events regulating the PM2.5-induced airway inflammation are yet to be elucidated. Considering the critical role of circular RNAs (circRNAs) in regulating inflammation, we predicted 11 circRNAs that may be involved in the PM2.5-induced airway inflammation using three previously reported miRNAs through the starBase website. A novel circRNA circ_0008553 was identified to be responsible for the PM2.5-activated inflammatory response in human bronchial epithelial cells (16HBE) via inducing oxidative stress. Using a combinatorial model PM2.5 library, we found that the synergistic effect of the insoluble core and loaded Zn2+ ions at environmentally relevant concentrations was the major contributor to the upregulation of circ_0008553 and subsequent induction of oxidative stress and inflammation in response to PM2.5 exposures. Our findings provided new insight into the intervention of PM2.5-induced adverse outcomes.


Asunto(s)
MicroARNs , ARN Circular , Células Epiteliales/metabolismo , Humanos , Inflamación/inducido químicamente , MicroARNs/metabolismo , Estrés Oxidativo , Material Particulado/toxicidad , Zinc/toxicidad
6.
Mol Biol Rep ; 49(9): 8575-8586, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35834034

RESUMEN

BACKGROUND: Pulpitis is a commonly seen oral inflammation condition in clinical practice, it can cause much pain for the patient and may induce infections in other systems. Much is still unknown for the pathogenic mechanism of pulpitis. In this work, we discovered that the expression of miR-155 was associated with dental pulpal inflammation both in vivo and in vitro. METHODS AND RESULTS: Our experiments of LPS stimulated odontoblast cell line MDPC-23 showed miR-155 could act as a positive regulator by increasing the production of pro-inflammatory cytokines IL-1ß and IL-6 during inflammatory responses, whereas knockdown of miR-155 can reverse the effects. Bioinformatics analysis demonstrated that SHIP1 is a direct target of miR-155 in odontoblasts, this result was further verified at both mRNA and protein level. Inhibition of miR-155 resulted in the downregulation of inflammation factors, while co-transfection of si-SHIP1 and miR-155 inhibitor promoted the inflammatory responses. Treatment with miR-155 mimic or si-SHIP1 up-regulated the protein level of p-PI3K and p-AKT. By contrast, miR-155 inhibitor exerted the opposite effects. miR-155 mimics could upregulate the gene expression of IL-1ß and IL-6. Co-transfection of LY294002 and miR-155 mimic attenuated the inflammatory responses. Consistent with in vitro results, miR-155-/- mice could alleviate inflammatory response, as well as decrease the activation of p-PI3K and p-AKT, whereas increase the activation of SHIP1. CONCLUSIONS: Our data revealed a novel role for miR-155 in regulation of dental pulpal inflammatory response by targeting SHIP1 through PI3K/AKT signaling pathway.


Asunto(s)
MicroARNs , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Pulpitis , Animales , Inflamación/genética , Interleucina-6/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pulpitis/genética
7.
Arch Toxicol ; 96(7): 2049-2065, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35435490

RESUMEN

Environmental chemical exposure often causes DNA damage, which leads to cellular dysfunction and the development of diseases. 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a tobacco-specific carcinogen that is known to cause DNA damage, while remains unknown about the underlying mechanism. In this study, simulated doses of NNK exposure in smokers, ranging from 50 to 300 µM, were used to detect the DNA damage effects of NNK in two human bronchial epithelial cells, 16HBE and BEAS-2B. The comet assay revealed increased DNA damage in response to NNK treatment, as measured by increased Olive tail moment (OTM). NNK treatment also led to elevated foci formation and protein expression of γ-H2AX, a DNA damage sensor. Dysregulation of proliferation, cell cycle arrest and apoptosis, was also observed in NNK-treated cells. Furthermore, the most effective dose of NNK (300 µM) was used in subsequent mechanistic studies. A circular RNA circNIPBL was identified to be significantly up-regulated in NNK-treated cells, circNIPBL knockdown successfully alleviated NNK-induced DNA damage and reversed the cellular dysregulation, while circNIPBL overexpression had the opposite effect. Mechanistically, we identified an interaction between circNIPBL and PARP1, a critical enzyme of the base excision repair (BER) pathway. CircNIPBL silencing successfully alleviated the NNK-induced inhibition of BER pathway proteins, including PARP1, XRCC1, PCNA and FEN1, while overexpression of circNIPBL had the opposite effect. In summary, our study shows for the first time that circNIPBL promotes NNK-induced DNA damage and cellular dysfunction through the BER pathway. In addition, our findings reveal the crucial role of epigenetic regulation in carcinogen-induced genetic lesions and further our understanding of environmental carcinogenesis.


Asunto(s)
Nitrosaminas , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Daño del ADN , Reparación del ADN , Epigénesis Genética , Células Epiteliales , Humanos , Nitrosaminas/toxicidad , ARN Circular , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
8.
Int J Med Sci ; 18(9): 1946-1952, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33850463

RESUMEN

Background: The world's first Diabetes Medications (Insulin) was marketed in October 1923. Some studies suggested the association of diabetes medications with Bullous Pemphigoid (BP), especially the Dipeptidyl Peptidase 4 (DPP-4) inhibitors. The study aims to detect an association between diabetes medications (focusing on DPP-4 inhibitors) and bullous pemphigoid based on FDA Adverse Event Reporting System (FAERS). Methods: All spontaneous reports of diabetes medications inhibitors-related BP recorded in the FAERS between March 2004 and August 2020 were included in the present study. Disproportionality analysis was performed to find the signal between diabetes medications and BP. The Chi-Squared with Yates' correction (χ2 Yates), proportional reporting ratio (PRR) and the lower limit of the 95% confidence interval of the Reporting Odds Ratio (ROR025) were calculated as a measure. A signal was detected when ROR025 > 1, PRR > 2, χ2 Yates > 4 and at least 3 cases. Results: There were 3770 reports for BP in FAERS. The strongest signal for diabetes medications-BP association were DDP-4 inhibitors (ROR025: 13.700, PRR: 15.408), followed by Meglitinides (ROR025: 12.708, PRR: 16.777), Non-sulfonylureas (ROR025: 6.434, PRR: 7.016), Alpha-glucosidase inhibitors (ROR025: 6.105, PRR: 10.738), Sulfonylureas (ROR025:2.655, PRR: 3.200). Conclusions: This study detected a strong signal between BP and DDP-4 inhibitors, alpha-glucosidase inhibitors, meglitinides, non-sulfonylureas, and sulfonylureas in FAERS. The signal was significantly higher with alogliptin than with the other DPP-4 inhibitors. The study doesn't suggest the association between the incretin mimetics, insulin, SGLT-2 inhibitors, thiazolidinediones and BP in FAERS.


Asunto(s)
Sistemas de Registro de Reacción Adversa a Medicamentos/estadística & datos numéricos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/efectos adversos , Penfigoide Ampolloso/epidemiología , Adulto , Anciano , Benzamidas/efectos adversos , Estudios de Casos y Controles , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Femenino , Inhibidores de Glicósido Hidrolasas/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Penfigoide Ampolloso/inducido químicamente , Farmacovigilancia , Estudios Retrospectivos , Compuestos de Sulfonilurea/efectos adversos , Estados Unidos/epidemiología , United States Food and Drug Administration/estadística & datos numéricos
9.
Arch Toxicol ; 95(10): 3235-3251, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34402960

RESUMEN

Fine particulate matter (PM2.5) has been shown to induce DNA damage. Circular RNAs (circRNAs) have been implicated in various disease processes related to environmental chemical exposure. However, the role of circRNAs in the regulation of DNA damage response (DDR) after PM2.5 exposure remains unclear. In this study, male ICR mice were exposed to PM2.5 at a daily mean concentration of 382.18 µg/m3 for 3 months in an enriched-ambient PM2.5 exposure system in Shijiazhuang, China, and PM2.5 collected form Shijiazhuang was applied to RAW264.7 cells at 100 µg/mL for 48 h. The results indicated that exposure to PM2.5 induced histopathological changes and DNA damage in the lung, kidney and spleen of male ICR mice, and led to decreased cell viability, increased LDH activity and DNA damage in RAW264.7 cells. Furthermore, circ_Cabin1 expression was significantly upregulated in multiple mouse organs as well as in RAW264.7 cells upon exposure to PM2.5. PM2.5 exposure also resulted in impairment of non-homologous end joining (NHEJ) repair via the downregulation of Lig4 or Dclre1c expression in vivo and in vitro. Importantly, circ_Cabin1 promoted PM2.5-induced DNA damage via inhibiting of NHEJ repair. Moreover, the expression of circ_Cabin1 and Lig4 or Dclre1c was strongly correlated in multiple mouse organs, as well as in the blood. In summary, our study provides a new perspective on circRNAs in the regulation of DDR after environmental chemical exposure.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Daño del ADN/efectos de los fármacos , Material Particulado/toxicidad , ARN Circular/genética , Animales , Supervivencia Celular/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/genética , ADN Ligasa (ATP)/genética , Endonucleasas/genética , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas Nucleares/genética , Células RAW 264.7
10.
Biophys J ; 119(8): 1606-1616, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32966761

RESUMEN

Activation of a gene is a multistep biochemical process, involving recruitments of transcription factors and histone kinases as well as modification of histones. Many of these intermediate reaction steps would have been unspecified by experiments. Therefore, classical two-state models of gene expression established based on the memoryless (or Markovian) assumption would not well describe the reality in gene expression. Recent experimental data have indicated that the inactive phases of gene promoters are differently distributed, showing strong memory. Here, we use a nonexponential waiting-time distribution to model the complex activation process of a gene, and then analyze a queuing model of stochastic transcription. We successfully derive the analytical expression of the stationary mRNA distribution, which provides insight into the effect of molecular memory created by complex activating events on the mRNA expression. We find that the reduction in the waiting-time noise may result in the increase in the mRNA noise, contrary to the previous conclusion. Based on the derived distribution, we also develop a method to infer the waiting-time distribution from a known mRNA distribution. Data analysis on a realistic example verifies the validity of this method.


Asunto(s)
Fenómenos Bioquímicos , Expresión Génica , Modelos Genéticos , Regulación de la Expresión Génica , ARN Mensajero/metabolismo , Procesos Estocásticos
11.
Mol Cancer ; 19(1): 101, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493389

RESUMEN

BACKGROUND: Lung cancer has high morbidity and mortality worldwide with non-small cell lung cancer (NSCLC) accounting for 85% of the cases. Therapies for lung cancer have relatively poor outcomes and further improvements are required. Circular RNAs have been reported to participate in the occurrence and progression of cancer. Information on the functions and mechanism of circRNAs in lung cancer is limited and needs more exploration. METHODS: We detected expression of genes and proteins by qPCR and western blot. Function of circSATB2 was investigated using RNA interference and overexpression assays. Location of circSATB2 was assessed by fluorescence in situ hybridization (FISH). Interaction of circSATB2, miR-326 and FSCN1 was confirmed by dual-luciferase reporter assay. RESULTS: Data from the investigation showed that circSATB2 was highly expressed in NSCLC cells and tissues. circSATB2 positively regulated fascin homolog 1, actin-bundling protein 1 (FSCN1) expression via miR-326 in lung cancer cells. Furthermore, circSATB2 can be transferred by exosomes and promote the proliferation, migration and invasion of NSCLC cells, as well as induce abnormal proliferation in normal human bronchial epithelial cells. Also, circSATB2 was highly expressed in serumal exosomes from lung cancer patients with high sensitivity and specificity for clinical detection and was related to lung cancer metastasis. CONCLUSIONS: circSATB2 participated in the progression of NSCLC and was differentially expressed in lung cancer tissue and serumal exosomes. circSATB2 may be potential biomarker for the diagnosis of NSCLC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Portadoras/metabolismo , Neoplasias Pulmonares/patología , Proteínas de Unión a la Región de Fijación a la Matriz/genética , MicroARNs/genética , Proteínas de Microfilamentos/metabolismo , ARN Circular/genética , Factores de Transcripción/genética , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Portadoras/genética , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas de Microfilamentos/genética , Pronóstico , Células Tumorales Cultivadas
12.
Mol Cancer ; 18(1): 13, 2019 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-30665425

RESUMEN

Circular RNAs are widely expressed in eukaryotic cells and associated with cancer. However, limited studies to date have focused on the potential role of circRNAs in progression of lung cancer. Data from the current investigation showed that circRNA 100146 is highly expressed in non-small cell lung cancer (NSCLC) cell lines and the chemically induced malignant transformed bronchial cell line, 16HBE-T, as well as 40 paired tissue samples of NSCLC. Suppression of circRNA 100146 inhibited the proliferation and invasion of cells and promoted apoptosis. Furthermore, circRNA 100146 could interact with splicing factors and bind miR-361-3p and miR-615-5p to regulate multiple downstream mRNAs. Our collective findings support a role of circRNA 100146 in the development of NSCLC and further demonstrate endogenous competition among circRNA 100146, SF3B3 and miRNAs, providing novel insights into the mechanisms underlying non-small cell lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Oncogenes , ARN/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , MicroARNs/metabolismo , ARN/metabolismo , ARN Circular , ARN Mensajero/genética
13.
Environ Sci Technol ; 53(8): 4566-4578, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30913382

RESUMEN

Airborne particular matter (PM2.5) contains complex mixtures of pollutants, and their compositions also vary with time and location. Inhalation of PM2.5 may cause a number of diseases, such as bronchial and lung inflammation and lung cancer. So far, how different components of PM2.5 contribute to inflammation and toxicity is still not known. To identify key PM2.5 components that are responsible for inflammation, here we took a reductionism approach and synthesized a model PM2.5 library containing 20 carbon nanoparticle based members with loadings of As(III), Pb2+, Cr(VI), and BaP individually or in combination at environment relevant concentrations. We discovered that only carbon nanoparticle-Pb2+ adducts, not other pollutants or adducts, induced inflammation in human bronchial cells by suppressing the expression of a novel long noncoding RNA lnc-PCK1-2:1, while lnc-PCK1-2:1 routinely plays a regulatory role in inhibiting inflammation. This finding was further substantiated by varying Pb2+ loadings on carbon nanoparticles and overexpressing lnc-PCK1-2:1. The success of this approach opens an avenue for further elucidation of molecular mechanisms of PM2.5-induced inflammation and toxicity.


Asunto(s)
Contaminantes Atmosféricos , ARN Largo no Codificante , Regulación hacia Abajo , Células Epiteliales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Plomo , Material Particulado , Fosfoenolpiruvato Carboxiquinasa (GTP)
14.
Carcinogenesis ; 39(8): 981-992, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-29726904

RESUMEN

Circular RNAs (circRNAs) are widespread and diverse endogenous RNAs distinct from traditional linear RNAs, which may regulate gene expression in eukaryotes. However, the function of human circRNAs, including their potential role in lung cancer, remains largely unknown. We screened the circRNA circ0006916, which was evidently down-regulated in 16HBE-T cells (anti-benzopyrene-trans-7, 8-dihydrodiol-9, 10-epoxide-transformed human bronchial epithelial cells), and in A549 and H460 cell lines. Silencing of circ0006916, but not its parental gene homer scaffolding protein 1 (HOMER1), promoted cell proliferation via speeding up the cell cycle process rather than by inhibiting apoptosis; conversely, overexpression of circ0006916 had the opposite effect. Luciferase-screening assay indicated that circ0006916 bound to miR-522-3p and inhibited pleckstrin homology domain and leucine rich repeat protein phosphatase 1 (PHLPP1) activity. We also explored the effect of the RNA-binding protein trinucleotide repeat-containing 6A (TNRC6A) on circ0006916 production. Circ0006916 expression was decreased after silencing TNRC6A. TNRC6A bound to the intron regions around the circRNA-forming exons of circ0006916, as shown by RNA immunoprecipitation assay combined with sequencing analysis. The association of circ0006916 with TNRC6A was further verified by RNA pull-down assays. We then constructed a carrier and confirmed that TNRC6A binding to the flanked intron region of circ0006916 was necessary for generation of circ0006916. These results demonstrate that TNRC6A regulates the biogenesis of the circRNA circ0006916, which has a regulatory role in cell growth.


Asunto(s)
Autoantígenos/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Células A549 , Autoantígenos/genética , Silenciador del Gen , Proteínas de Andamiaje Homer/genética , Humanos , Neoplasias Pulmonares/patología , MicroARNs/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , ARN/genética , ARN Circular , Proteínas de Unión al ARN/genética
15.
Toxicol Appl Pharmacol ; 342: 22-30, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29391238

RESUMEN

While hexavalent chromium [Cr(VI)] is generally considered as a genotoxic environmental carcinogen, studies showed that Cr(VI) exposure also causes epigenetic changes. However, whether Cr(VI)-caused epigenetic dysregulations plays an important role in Cr(VI) carcinogenicity remain largely unknown. The aim of this study was to determine if chronic low dose Cr(VI) exposure causes epigenetic changes, the underlying mechanism and whether chronic low dose Cr(VI) exposure-caused epigenetic dysregulation contributes causally to Cr(VI)-induced cancer stem cell (CSC)-like property and cell transformation. Two immortalized human bronchial epithelial cell lines (BEAS-2B and 16HBE) were exposed to 0.25 µM of K2Cr2O7 for 20 and 40 weeks to induce cell transformation, respectively. Cr(VI)-induced epigenetic changes were examined in Cr(VI)-transformed cells and Cr(VI) exposure-caused human lung cancer tissues. Pharmacological inhibitors and gene knockdown experiments were used to determine the role of epigenetic dysregulation in Cr(VI) carcinogenicity. We found that chronic Cr(VI) exposure causes epigenetic dysregulation as evidenced by the increased levels of histone H3 repressive methylation marks (H3K9me2 and H3K27me3) and the related histone-lysing methyltransferases (HMTases). Pharmacological inhibition or knockdown of HMTases reduces H3 repressive methylation marks and malignant phenotypes of Cr(VI)-transformed cells. Moreover, knockdown of HMTases in parental cells significantly reduces chronic Cr(VI) exposure-induced CSC-like property and cell transformation. Further mechanistic study revealed that knockdown of HMTases decreases Cr(VI) exposure-caused DNA damage. Our findings indicate that chronic Cr(VI) exposure increases H3 repressive methylation marks by increasing the related HMTases expression; and that increased expression of HMTases plays a causal role in Cr(VI)-induced CSC-like property and cell transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Cromo/toxicidad , N-Metiltransferasa de Histona-Lisina/biosíntesis , Células Madre Neoplásicas/metabolismo , Regulación hacia Arriba/fisiología , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
16.
Ecotoxicol Environ Saf ; 157: 380-387, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29635186

RESUMEN

Air pollution worldwide, especially in China and India, has caused serious health issues. Because PM2.5 particles consist of solid particles of diverse properties with payloads of inorganic, organic and biological pollutants, it is still not known what the major toxic components are and how these components induce toxicities. To explore this complex issue, we apply reductionism principle and an ultrafine particle library approach in this work. From investigation of 63 diversely functionalized ultrafine particles (FUPs) with adsorbed key pollutants, our findings indicate that 1) only certain pollutants in the payloads of PM2.5 are responsible for causing cellular oxidative stress, cell apoptosis, and cytotoxicity while the particle carriers are much less toxic; 2) pollutant-induced cellular oxidative stress and oxidative stress-triggered apoptosis are identified as one of the dominant mechanisms for PM2.5-induced cytotoxicity; 3) each specific toxic component on PM2.5 (such as As, Pb, Cr or BaP) mainly affects its specific target organ(s) and, adding together, these pollutants may cause synergistic or just additive effects. Our findings demonstrate that reductionism concept and model PM2.5 particle library approach are very effective in our endeavor to search for a better understanding of PM2.5-induced health effects.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Apoptosis , Estrés Oxidativo , Material Particulado/toxicidad , Contaminación del Aire/efectos adversos , Bronquios/citología , Células Cultivadas , China , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células HEK293 , Humanos , India , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo
17.
Environ Sci Technol ; 51(16): 9334-9343, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28723108

RESUMEN

As the applications and environmental release of silver ions and nanoparticles are increasing, increasing human exposure to these pollutants has become an emerging health concern. The impeding effects of such pollutants on susceptible populations are severely under-studied. Here, we demonstrate that silver nanoparticles (Ag NPs), at a dose that causes no general toxicity in normal mice, promotes the progression of fatty liver disease from steatosis to steatohepatitis only in overweight mice. Exposure to Ag+ ions induces the same effects in overweight mice. Ag NPs rather than Ag+ ions cause this disease progression based on our findings that Ag+ ions are partly reduced to Ag NPs in fatty livers, and the toxic effect is correlated with the liver dose of Ag NPs, not Ag+ ions. Furthermore, the Ag NP-induced pro-inflammatory activation of Kupffer cells in the liver, enhancement of hepatic inflammation, and suppression of fatty acid oxidation are identified as key factors in the underlying mechanisms.


Asunto(s)
Nanopartículas del Metal/toxicidad , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Sobrepeso , Plata/toxicidad , Animales , Iones , Hepatopatías , Ratones , Estrés Oxidativo
18.
Arch Toxicol ; 91(4): 1671-1684, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27604105

RESUMEN

Lead is a metal that has toxic effects on the developing nervous system. However, the mechanisms underlying lead-induced neurotoxicity are not well understood. Non-coding RNAs (ncRNAs) play an important role in epigenetic regulation, but few studies have examined the function of ncRNAs in lead-induced neurotoxicity. We addressed this in the present study by evaluating the functions of a long non-coding RNA (named lncRpa) and a circular RNA (named circRar1) in a mouse model of lead-induced neurotoxicity. High-throughput RNA sequencing showed that both lncRpa and circRar1 promoted neuronal apoptosis. We also found that lncRpa and circRar1 induced the upregulation of apoptosis-associated factors caspase8 and p38 at the mRNA and protein levels via modulation of their common target microRNA miR-671. This is the first report of a regulatory interaction among a lncRNA, circRNA, and miRNA mediating neuronal apoptosis in response to lead toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Compuestos Organometálicos/toxicidad , Animales , Apoptosis/genética , Caspasa 8/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Ratones , MicroARNs/genética , Neuroblastoma/genética , Síndromes de Neurotoxicidad/genética , ARN/genética , ARN Circular , ARN Largo no Codificante/genética , Regulación hacia Arriba/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
19.
J Biol Chem ; 289(26): 18373-86, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24841200

RESUMEN

MicroRNA-200b (miR-200b) is a member of miR-200 family that has been found to inhibit cell migration and cancer metastasis; however, the underlying mechanism is not well understood. We previously reported that miR-200 expression is depleted in arsenic-transformed human bronchial epithelial cells with highly migratory and invasive characteristics, whereas stably re-expressing miR-200b strongly suppresses arsenic-transformed cell migration. This study was performed to investigate how miR-200b inhibits arsenic-transformed cell migration. We found that protein kinase Cα (PKCα) is significantly up-regulated in arsenic-transformed cells. Combining bioinformatics analysis with PKCα 3'-untranslated region vector luciferase reporter assays, we showed that PKCα is a direct target of miR-200b. Inhibiting PKCα activity or knocking down PKCα expression drastically reduced cell migration, phenocoping the inhibitory effect of overexpressing miR-200b. In contrast, forced expression of PKCα in miR-200b overexpressing cells impaired the inhibitory effect of miR-200b on cell migration. In addition, we also found a positive feedback loop between Wnt5b and PKCα in arsenic-transformed cells. Knocking down Wnt5b expression reduced phospho-PKC levels and cell migration; and knocking down PKCα expression decreased Wnt5b level and cell migration. Moreover, forced expression of PKCα increased Wnt5b and phospho-PKC levels and cell migration. Further mechanistic studies revealed that Rac1 is highly activated in arsenic-transformed cells and stably expressing miR-200b abolishes Rac1 activation changing actin cytoskeleton organization. Manipulating PKCα or Wnt5b expression levels significantly altered the level of active Rac1. Together, these findings indicate that miR-200b suppresses arsenic-transformed cell migration by targeting PKCα and Wnt5b-PKCα positive feedback loop and subsequently inhibiting Rac1 activation.


Asunto(s)
Arsénico/toxicidad , Movimiento Celular , MicroARNs/metabolismo , Proteína Quinasa C-alfa/genética , Proteínas Wnt/genética , Proteína de Unión al GTP rac1/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática , Retroalimentación Fisiológica , Humanos , MicroARNs/genética , Unión Proteica , Proteína Quinasa C-alfa/metabolismo , Proteínas Wnt/metabolismo , Proteína de Unión al GTP rac1/genética
20.
Mol Carcinog ; 54 Suppl 1: E192-204, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25820656

RESUMEN

Lung cancer is a major health problem, and is considered one of the deadliest cancers in humans. It is refractory to current treatments, and the mechanisms of lung cancer are unknown. Long noncoding RNAs (lncRNAs) are involved in various biological processes and human diseases. However, the exact functional roles and mechanisms of lncRNAs are largely unclear. In this study, we attempted to identify lung-cancer-related lncRNAs. We found changes in lncRNA expression in the anti-benzo(a) pyrene-7,8-diol-9,10-epoxide (anti-BPDE)-transformed human bronchial epithelial cell line (16HBE-T cells) using microarrays and qRT-PCR. Of these lncRNAs, LOC728228 was upregulated relative to its expression in control untransformed16HBE (16HBE-N) cells. LOC728228 knockdown inhibited cell proliferation, caused G0/G1-phase cell-cycle arrest, reduced cellular migration, suppressed colony formation in vitro, and inhibited tumor growth in a nude mouse xenograft model. LOC728228 knockdown also suppressed cyclin D1 expression, and the depletion of cyclin D1 induced G0/G1-phase cell-cycle arrest and inhibited cell proliferation, thus influencing the malignant potential of cancer cells. In summary, our results suggest that lncRNA LOC728228 has an oncogene-like function and plays a vital role in human lung cancer.


Asunto(s)
7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/toxicidad , Neoplasias Pulmonares/patología , ARN Largo no Codificante/fisiología , Animales , Línea Celular Transformada , Humanos , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA