Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Acta Pharmacol Sin ; 45(7): 1492-1505, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38538718

RESUMEN

Immunosuppression by the tumor microenvironment is a pivotal factor contributing to tumor progression and immunotherapy resistance. Priming the tumor immune microenvironment (TIME) has emerged as a promising strategy for improving the efficacy of cancer immunotherapy. In this study we investigated the effects of noninvasive radiofrequency radiation (RFR) exposure on tumor progression and TIME phenotype, as well as the antitumor potential of PD-1 blockage in a model of pulmonary metastatic melanoma (PMM). Mouse model of PMM was established by tail vein injection of B16F10 cells. From day 3 after injection, the mice were exposed to RFR at an average specific absorption rate of 9.7 W/kg for 1 h per day for 14 days. After RFR exposure, lung tissues were harvested and RNAs were extracted for transcriptome sequencing; PMM-infiltrating immune cells were isolated for single-cell RNA-seq analysis. We showed that RFR exposure significantly impeded PMM progression accompanied by remodeled TIME of PMM via altering the proportion and transcription profile of tumor-infiltrating immune cells. RFR exposure increased the activation and cytotoxicity signatures of tumor-infiltrating CD8+ T cells, particularly in the early activation subset with upregulated genes associated with T cell cytotoxicity. The PD-1 checkpoint pathway was upregulated by RFR exposure in CD8+ T cells. RFR exposure also augmented NK cell subsets with increased cytotoxic characteristics in PMM. RFR exposure enhanced the effector function of tumor-infiltrating CD8+ T cells and NK cells, evidenced by increased expression of cytotoxic molecules. RFR-induced inhibition of PMM growth was mediated by RFR-activated CD8+ T cells and NK cells. We conclude that noninvasive RFR exposure induces antitumor remodeling of the TIME, leading to inhibition of tumor progression, which provides a promising novel strategy for TIME priming and potential combination with cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Células Asesinas Naturales , Neoplasias Pulmonares , Ratones Endogámicos C57BL , Microambiente Tumoral , Animales , Células Asesinas Naturales/inmunología , Microambiente Tumoral/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Linfocitos T CD8-positivos/inmunología , Ratones , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Fenotipo , Receptor de Muerte Celular Programada 1 , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología
2.
Ecotoxicol Environ Saf ; 223: 112554, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34332247

RESUMEN

Arsenic is one of the most common environmental pollutants. Neurotoxicity induced by arsenic has become a major public health concern. However, the effects of arsenic-induced neurotoxicity in the brain and the underlying molecular mechanisms are not well understood. N-acetyl-cysteine (NAC) is a thiol-based antioxidant that can antagonize heavy metal-induced neurotoxicity by scavenging reactive oxygen species (ROS). Here, we used the mouse oligodendrocyte precursor cell (OPC) line Oli-neu to explore the neurotoxic effects of arsenic and the protective effects of NAC. We found that arsenic exposure decreased cell viability, increased oxidative stress, caused mitochondrial dysfunction, and led to apoptosis of Oli-neu cells. Furthermore, we revealed that NAC treatment reversed these neurotoxic effects of arsenic. TMEM179, a key membrane protein, was found highly expressed in OPCs and to be an important factor in maintaining mitochondrial functions. We found that TMEM179 played a critical role in mediating the neurotoxic effects of arsenic and the protective role of NAC. PKCß is a downstream factor through which TMEM179 regulates the expression of apoptosis-related proteins. This study improves our understanding of the neurotoxic effects and mechanisms of arsenic exposure and the protective effects of NAC. It also identifies a potential molecular target, TMEM179, for the treatment of arsenic-induced neurotoxicity.


Asunto(s)
Acetilcisteína , Arsénico , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Animales , Apoptosis , Arsénico/metabolismo , Arsénico/toxicidad , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
3.
Cell Tissue Res ; 368(1): 145-157, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27807703

RESUMEN

Toll-like receptor 4 (TLR4) plays critical roles in vascular inflammation, lipid accumulation and atherosclerosis development. However, the mechanisms underlying these processes are still not well established, especially in vascular smooth muscle cells (VSMCs). ATP-binding cassette transporter G1 (ABCG1) is one of the key genes mediating inflammation and cellular lipid accumulation. The function of TLR4 in regulating the expression of ABCG1 and the underlying molecular mechanisms remain to be elucidated. In this study, we cultured VSMCs from the thoracic aortas of mice and treated the cells with 50 µg/ml oxidized low-density lipoprotein (oxLDL) to activate TLR4 signaling. We observed that activating TLR4 with oxLDL induced inflammatory responses and lipid accumulation in VSMCs. The expression of peroxisome proliferator-activated receptor gamma (PPARγ), liver X receptor alpha (LXRα) and ABCG1 was inhibited by TLR4 activation. However, these effects could be reversed by knocking out TLR4. PPARγ activation by rosiglitazone rescued LXRα and ABCG1 expression and reduced TLR4-induced inflammation and lipid accumulation. Silencing PPARγ expression with a specific small interfering RNA (siRNA) inhibited LXRα and ABCG1 expression and, importantly, enhanced TLR4-induced inflammation and lipid accumulation. In conclusion, ABCG1 expression was down-regulated by TLR4, which induces inflammation and lipid accumulation in VSMCs via PPARγ/LXRα signaling. These findings indicate a novel molecular mechanism underlying TLR4-induced inflammation and lipid accumulation.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Receptores X del Hígado/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , PPAR gamma/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas LDL/farmacología , Ratones Noqueados , Modelos Biológicos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Receptor Toll-Like 4/deficiencia
4.
J Pineal Res ; 62(3)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28099758

RESUMEN

Cadmium (Cd) is a persistent environmental and occupational contaminant that accumulates in the liver and induces oxidative stress and inflammation. Melatonin possesses potent hepatoprotective properties against the development and progression of acute and chronic liver injury. Nevertheless, the molecular mechanism underlying the protective effects of melatonin against Cd-induced hepatotoxicity remains obscure. In this study, we aimed to investigate the effects of melatonin on Cd-induced liver inflammation and hepatocyte death. Male C57BL/6 mice were intraperitoneally injected with melatonin (10 mg/kg) once a day for 3 days before exposure to CdCl2 (2.0 mg/kg). We found that Cd induced hepatocellular damage and inflammatory infiltration as well as increased serum ALT/AST enzymes. In addition, we showed that Cd triggered an inflammatory cell death, which is mediated by the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Moreover, melatonin treatment significantly alleviated Cd-induced liver injury by decreasing serum ALT/AST levels, suppressing pro-inflammatory cytokine production, inhibiting NLRP3 inflammasome activation, ameliorating oxidative stress, and attenuating hepatocyte death. Most importantly, melatonin markedly abrogated Cd-induced TXNIP overexpression and decreased the interaction between TXNIP and NLRP3 in vivo and in vitro. However, treatment with siRNA targeting TXNIP blocked the protective effects of melatonin in Cd-treated primary hepatocytes. Collectively, our results suggest that melatonin confers protection against Cd-induced liver inflammation and hepatocyte death via inhibition of the TXNIP-NLRP3 inflammasome pathway.


Asunto(s)
Cadmio/toxicidad , Proteínas Portadoras , Enfermedad Hepática Inducida por Sustancias y Drogas , Inflamasomas , Melatonina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR , Tiorredoxinas , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Muerte Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Tiorredoxinas/biosíntesis , Tiorredoxinas/genética
5.
Cereb Cortex ; 26(9): 3690-3704, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27405333

RESUMEN

Glial precursor transplantation provides a potential therapy for brain disorders. Before its clinical application, experimental evidence needs to indicate that engrafted glial cells are functionally incorporated into the existing circuits and become essential partners of neurons for executing fundamental brain functions. While previous experiments supporting for their functional integration have been obtained under in vitro conditions using slice preparations, in vivo evidence for such integration is still lacking. Here, we utilized in vivo two-photon Ca(2+) imaging along with immunohistochemistry, fluorescent indicator labeling-based axon tracing and correlated light/electron microscopy to analyze the profiles and the functional status of glial precursor cell-derived astrocytes in adult mouse neocortex. We show that after being transplanted into somatosensory cortex, precursor-derived astrocytes are able to survive for more than a year and respond with Ca(2+) signals to sensory stimulation. These sensory-evoked responses are mediated by functionally-expressed nicotinic receptors and newly-established synaptic contacts with the host cholinergic afferents. Our results provide in vivo evidence for a functional integration of transplanted astrocytes into adult mammalian neocortex, representing a proof-of-principle for sensory cortex remodeling through addition of essential neural elements. Moreover, we provide strong support for the use of glial precursor transplantation to understand glia-related neural development in vivo.


Asunto(s)
Astrocitos/fisiología , Astrocitos/trasplante , Señalización del Calcio/fisiología , Estimulación Eléctrica/métodos , Neocórtex/citología , Neocórtex/fisiología , Corteza Somatosensorial/fisiología , Potenciales de Acción/fisiología , Animales , Astrocitos/citología , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Ratones , Neocórtex/cirugía , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Transducción de Señal/fisiología
6.
Commun Biol ; 7(1): 997, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39147853

RESUMEN

The effects of neurotoxicant cadmium (Cd) exposure on brain development have not been well elucidated. To investigate this, we have herein subjected pregnant mice to low-dose Cd throughout gestation. Using single-cell RNA sequencing (scRNA-seq), we explored the cellular responses in the embryonic brain to Cd exposure, and identified 18 distinct cell subpopulations that exhibited varied responses to Cd. Typically, Cd exposure impeded the development and maturation of cells in the brain, especially progenitor cells such as neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). It also caused significant cell subpopulation shifts in almost all the types of cells in the brain. Additionally, Cd exposure reduced the dendritic sophistication of cortical neurons in the offspring. Importantly, these changes led to aberrant Ca2+ activity in the cortex and neural behavior changes in mature offspring. These data contribute to our understanding of the effects and mechanisms of Cd exposure on brain development and highlight the importance of controlling environmental neurotoxicant exposure at the population level.


Asunto(s)
Encéfalo , Cadmio , Análisis de la Célula Individual , Transcriptoma , Animales , Ratones , Cadmio/toxicidad , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Femenino , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/efectos de los fármacos , Ratones Endogámicos C57BL , Masculino , Neuronas/metabolismo , Neuronas/efectos de los fármacos
7.
Adv Sci (Weinh) ; 11(30): e2402030, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38837686

RESUMEN

Cadmium (Cd) is a neurotoxic contaminant that induces cognitive decline similar to that observed in Alzheimer's disease (AD). Autophagic flux dysfunction is attributed to the pathogenesis of AD, and this study aimed to investigate the effect of autophagy on environmental Cd-induced AD progression and the underlying mechanism. Here, Cd exposure inhibited autophagosome-lysosome fusion and impaired lysosomal function, leading to defects in autophagic clearance and then to APP accumulation and nerve cell death. Proteomic analysis coupled with Ingenuity Pathway Analysis (IPA) identified SIRT5 as an essential molecular target in Cd-impaired autophagic flux. Mechanistically, Cd exposure hampered the expression of SIRT5, thus increasing the succinylation of RAB7A at lysine 31 and inhibiting RAB7A activity, which contributed to autophagic flux blockade. Importantly, SIRT5 overexpression led to the restoration of autophagic flux blockade, the alleviation of Aß deposition and memory deficits, and the desuccinylation of RAB7A in Cd-exposed FAD4T mice. Additionally, SIRT5 levels decrease mainly in neurons but not in other cell clusters in the brains of AD patients according to single-nucleus RNA sequencing data from the public dataset GSE188545. This study reveals that SIRT5-catalysed RAB7A desuccinylation is an essential adaptive mechanism for the amelioration of Cd-induced autophagic flux blockade and AD-like pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Autofagia , Cadmio , Modelos Animales de Enfermedad , Sirtuinas , Proteínas de Unión al GTP rab , Proteínas de Unión a GTP rab7 , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Animales , Ratones , Cadmio/metabolismo , Cadmio/toxicidad , Autofagia/efectos de los fármacos , Sirtuinas/metabolismo , Sirtuinas/genética , Proteínas de Unión a GTP rab7/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/genética , Humanos , Masculino
8.
Sci Total Environ ; 918: 170773, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38336054

RESUMEN

Cadmium (Cd) exposure is known to enhance breast cancer (BC) progression. Cd promotes epithelial-mesenchymal transition (EMT) in BC cells, facilitating BC cell aggressiveness and invasion, but the underlying molecular mechanisms are unclear. Hence, transgenic MMTV-Erbb2 mice (6 weeks) were orally administered Cd (3.6 mg/L, approximately equal to 19.64 µΜ) for 23 weeks, and BC cells (BT474 cells) were exposed to Cd (0, 0.1, 1 or 10 µΜ) for 72 h to investigate the effect of Cd exposure on EMT in BC cells. Chronic Cd exposure dramatically expedited tumor metastasis to multiple organs; decreased E-cadherin density; and increased Vimentin, N-cadherin, ZEB1, and Twist density in the tumor tissues of MMTV-Erbb2 mice. Notably, transcriptomic analysis of BC tumors revealed cytochrome P450 1B1 (CYP1B1) as a key factor that regulates EMT progression in Cd-treated MMTV-Erbb2 mice. Moreover, Cd increased CYP1B1 expression in MMTV-Erbb2 mouse BC tumors and in BT474 cells, and CYP1B1 inhibition decreased Cd-induced BC cell malignancy and EMT in BT474 cells. Importantly, the promotion of EMT by CYP1B1 in Cd-treated BC cells was presumably controlled by glutamine metabolism. This study offers novel perspectives into the effect of environmental Cd exposure on driving BC progression and metastasis, and this study provides important guidance for comprehensively assessing the ecological and health risks of Cd.


Asunto(s)
Cadmio , Neoplasias , Ratones , Animales , Cadmio/farmacología , Línea Celular Tumoral , Glutamina/metabolismo , Glutamina/farmacología , Reprogramación Metabólica , Transición Epitelial-Mesenquimal , Cadherinas/genética , Cadherinas/metabolismo , Cadherinas/farmacología
9.
iScience ; 26(10): 107418, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37771661

RESUMEN

Terahertz (THz) is an electromagnetic wave with a radiation wavelength range of 30-3000 µm and a frequency of 0.1-10 THz. With the development of new THz sources and devices, THz has been widely applied in various fields. However, there are few studies on biological effects of THz irradiation on the human neural stem cells (hNSCs) and mouse neural stem cells (mNSCs), which need to be further studied. We studied the biological effects of THz radiation on hNSCs and mNSCs. The effects of THz irradiation time and average output power on the proliferation, apoptosis, and DNA damage of NSCs were analyzed by flow cytometry and immunofluorescence. The results showed that the proliferation and apoptosis of NSCs were dose-dependently affected by THz irradiation time and average output power. The proliferation of hNSCs was more vulnerable to damage and apoptosis was more serious under the same terahertz irradiation conditions compared to those of mNSCs.

10.
Sci Total Environ ; 905: 167039, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37716689

RESUMEN

Cadmium (Cd), a predominant environmental pollutant, is a canonical toxicant that acts on the kidneys. However, the nephrotoxic effect and underlying mechanism activated by chronic exposure to Cd remain unclear. In the present study, male mice (C57BL/6J, 8 weeks) were treated with 0.6 mg/L cadmium chloride (CdCl2) administered orally for 6 months, and tubular epithelial cells (TCMK-1 cells) were treated with low-dose (1, 2, and 3 µM) CdCl2 for 72 h (h). Our study results revealed that environmental Cd exposure triggered ferroptosis and renal dysfunction. Spatially resolved metabolomics enabled delineation of metabolic profiles and visualization of the disruption to glutathione homeostasis related to ferroptosis in mouse kidneys. Multiomics analysis revealed that chronic Cd exposure induced glutathione redox imbalance that depended on STEAP3-driven lysosomal iron overload. In particular, glutathione metabolic reprogramming linked to ferroptosis emerged as a metabolic hallmark in the blood of Cd-exposed workers. In conclusion, this study provides the first evidence indicating that chronic Cd exposure triggers ferroptosis and renal dysfunction that depend on STEAP3-mediated glutathione redox imbalance, greatly increasing our understanding of the metabolic reprogramming induced by Cd exposure in the kidneys and providing novel clues linking chronic Cd exposure to nephrotoxicity.


Asunto(s)
Ferroptosis , Enfermedades Renales , Humanos , Masculino , Ratones , Animales , Cadmio/toxicidad , Cadmio/metabolismo , Ratones Endogámicos C57BL , Oxidación-Reducción , Enfermedades Renales/inducido químicamente , Glutatión/metabolismo
11.
Stem Cell Res Ther ; 13(1): 285, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35765112

RESUMEN

Aging causes astrocyte morphological degeneration and functional deficiency, which impairs neuronal functions. Until now, whether age-induced neuronal deficiency could be alleviated by engraftment of glial progenitor cell (GPC) derived astrocytes remained unknown. In the current study, GPCs were generated from embryonic cortical neural stem cells in vitro and transplanted into the brains of aged mice. Their integration and intervention effects in the aged brain were examined 12 months after transplantation. Results indicated that these in-vitro-generated GPC-derived astrocytes possessed normal functional properties. After transplantation they could migrate, differentiate, achieve long-term integration, and maintain much younger morphology in the aged brain. Additionally, these GPC-derived astrocytes established endfeet expressing aquaporin-4 (AQP4) and ameliorate AQP4 polarization in the aged neocortex. More importantly, age-dependent sensory response degeneration was reversed by GPC transplantation. This work demonstrates that rejuvenation of the astrocyte niche is a promising treatment to prevent age-induced degradation of neuronal and behavioral functions.


Asunto(s)
Células-Madre Neurales , Neuroglía , Animales , Astrocitos/metabolismo , Ratones , Neuronas , Trasplante de Células Madre
12.
J Inorg Biochem ; 234: 111901, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35716551

RESUMEN

Cadmium (Cd), a common environmental and occupational toxicant, is an important risk factor for hearing loss. After exposure, Cd accumulates in the inner ear and induces spiral ganglion neuron (SGN) degeneration; however, the underlying mechanisms are poorly understood. Dysfunctional autophagy has been implicated in many neurodegenerative diseases, including Cd-induced neurotoxicity. Metformin has been validated to confer not only anti-hyperglycaemic but also neuroprotective effects. However, the relationship between autophagy dysfunction, SGN degeneration, and the effect of metformin on Cd-induced SGN neurotoxicity has not yet been established. In this study, we demonstrate that metformin notably attenuates Cd-evoked SGN degeneration by restoring impaired autophagy flux, as evidenced by the suppression of Cd-induced elevation of autophagy markers microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and autophagy substrate protein p62 in degenerated SGN. Blockage of autophagy flux by chloroquine abolished metformin-induced neuroprotection against Cd-induced neurotoxicity in SGN. The results of this study reveal that autophagy dysfunction is an important component of Cd-induced SGN degeneration, and metformin may be a potential protective agent for attenuating SGN degeneration following Cd exposure.


Asunto(s)
Cadmio , Metformina , Autofagia , Cadmio/metabolismo , Metformina/metabolismo , Metformina/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Ganglio Espiral de la Cóclea/metabolismo
13.
J Hazard Mater ; 435: 128942, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35468398

RESUMEN

Metabolic disorders induced by arsenic exposure have attracted great public concern. However, it remains unclear whether hypothalamus-based central regulation mechanisms are involved in this process. Here, we exposed mice to 100 µg/L arsenic in drinking water and established a chronic arsenic exposure model. Our study revealed that chronic arsenic exposure caused metabolic disorders in mice including impaired glucose metabolism and decreased energy expenditure. Arsenic exposure also impaired glucose sensing and the activation of proopiomelanocortin (POMC) neurons in the hypothalamus. In particular, arsenic exposure damaged the plasticity of hypothalamic astrocytic process. Further research revealed that arsenic exposure inhibited the expression of sex-determining region Y-Box 2 (SOX2), which decreased the expression level of insulin receptors (INSRs) and the phosphorylation of AKT. The conditional deletion of astrocytic SOX2 exacerbated arsenic-induced effects on metabolic disorders, the impairment of hypothalamic astrocytic processes, and the inhibition of INSR/AKT signaling. Furthermore, the arsenic-induced impairment of astrocytic processes and inhibitory effects on INSR/AKT signaling were reversed by SOX2 overexpression in primary hypothalamic astrocytes. Together, we demonstrated here that chronic arsenic exposure caused metabolic disorders by impairing SOX2-modulated hypothalamic astrocytic process plasticity in mice. Our study provides evidence of novel central regulatory mechanisms underlying arsenic-induced metabolic disorders and emphasizes the crucial role of SOX2 in regulating the process plasticity of adult astrocytes.


Asunto(s)
Arsénico , Enfermedades Metabólicas , Animales , Arsénico/metabolismo , Arsénico/toxicidad , Hipotálamo/metabolismo , Enfermedades Metabólicas/metabolismo , Ratones , Proopiomelanocortina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
Front Public Health ; 9: 771508, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34881219

RESUMEN

Background: With the global popularity of communication devices such as mobile phones, there are increasing concerns regarding the effect of radiofrequency electromagnetic radiation (RF-EMR) on the brain, one of the most important organs sensitive to RF-EMR exposure at 1,800 MHz. However, the effects of RF-EMR exposure on neuronal cells are unclear. Neurite outgrowth plays a critical role in brain development, therefore, determining the effects of 1,800 MHz RF-EMR exposure on neurite outgrowth is important for exploring its effects on brain development. Objectives: We aimed to investigate the effects of 1,800 MHz RF-EMR exposure for 48 h on neurite outgrowth in neuronal cells and to explore the associated role of the Rap1 signaling pathway. Material and Methods: Primary hippocampal neurons from C57BL/6 mice and Neuro2a cells were exposed to 1,800 MHz RF-EMR at a specific absorption rate (SAR) value of 4 W/kg for 48 h. CCK-8 assays were used to determine the cell viability after 24, 48, and 72 h of irradiation. Neurite outgrowth of primary hippocampal neurons (DIV 2) and Neuro2a cells was observed with a 20 × optical microscope and recognized by ImageJ software. Rap1a and Rap1b gene expressions were detected by real-time quantitative PCR. Rap1, Rap1a, Rap1b, Rap1GAP, and p-MEK1/2 protein expressions were detected by western blot. Rap1-GTP expression was detected by immunoprecipitation. The role of Rap1-GTP was assessed by transfecting a constitutively active mutant plasmid (Rap1-Gly_Val-GFP) into Neuro2a cells. Results: Exposure to 1,800 MHz RF-EMR for 24, 48, and 72 h at 4 W/kg did not influence cell viability. The neurite length, primary and secondary neurite numbers, and branch points of primary mouse hippocampal neurons were significantly impaired by 48-h RF-EMR exposure. The neurite-bearing cell percentage and neurite length of Neuro2a cells were also inhibited by 48-h RF-EMR exposure. Rap1 activity was inhibited by 48-h RF-EMR with no detectable alteration in either gene or protein expression of Rap1. The protein expression of Rap1GAP increased after 48-h RF-EMR exposure, while the expression of p-MEK1/2 protein decreased. Overexpression of constitutively active Rap1 reversed the decrease in Rap1-GTP and the neurite outgrowth impairment in Neuro2a cells induced by 1,800 MHz RF-EMR exposure for 48 h. Conclusion: Rap1 activity and related signaling pathways are involved in the disturbance of neurite outgrowth induced by 48-h 1,800 MHz RF-EMR exposure. The effects of RF-EMR exposure on neuronal development in infants and children deserve greater focus.


Asunto(s)
Hipocampo , Neuronas , Animales , Radiación Electromagnética , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/farmacología , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Humanos , Ratones , Ratones Endogámicos C57BL , Proyección Neuronal , Neuronas/metabolismo , Neuronas/efectos de la radiación
15.
J Hazard Mater ; 402: 123926, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33254826

RESUMEN

Bisphenol A (BPA), an environmental endocrine-disrupting compound, has been revealed associated with metabolic disorders such as obesity, prediabetes, and type 2 diabetes (T2D). However, its underlying mechanisms are still not fully understood. Here, we provide new evidence that BPA is a risk factor for T2D from a case-control study. To explore the detailed mechanisms, we used two types of diet models, standard diet (SD) and high-fat diet (HFD), to study the effects of long-term BPA exposure on prediabetes in 4-week-old mice. We found that BPA exposure for 12 weeks exacerbated HFD-induced prediabetic symptoms. Female mice showed increased body mass, serum insulin level, and impaired glucose tolerance, while male mice only exhibited impaired glucose tolerance. No change was found in SD-fed mice. Besides, BPA exposure enhanced astrocyte-dependent hypothalamic inflammation in both male and female mice, which impaired proopiomelanocortin (POMC) neuron functions. Moreover, eliminating inflammation by toll-like receptor 4 (TLR4) knockout significantly abolished the effects of BPA on the hypothalamus and diet-induced prediabetes. Taken together, our data establish a key role for TLR4-dependent hypothalamic inflammation in regulating the effects of BPA on prediabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Estado Prediabético , Animales , Compuestos de Bencidrilo/toxicidad , Estudios de Casos y Controles , Femenino , Hipotálamo/metabolismo , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Fenoles , Estado Prediabético/inducido químicamente , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
16.
Chemosphere ; 268: 129221, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33352510

RESUMEN

Bisphenol A (BPA) is a high-production-volume industrial chemical that facilitates the development of breast cancer. However, the molecular mechanism associated with BPA-induced breast cancer cell proliferation and migration remains elusive. In our study, we exposed MCF-7 cells to different concentrations of BPA (0.1, 1 and 10 µM) for 24, 48, or 72 h. We found that BPA exposure significantly promoted MCF-7 cell proliferation and migration but not invasion. To elucidate the mechanisms, the differentially expressed genes between the BPA and control groups were investigated with the Gene Expression Omnibus (GEO) database through GEO2R. Kyoto Encyclopedia of Genes and Genomes (KEGG) and pathway action network analyses demonstrated the important role of the cell cycle pathway in the effects of BPA exposure on MCF-7 cells. Importantly, analysis with the cytoHubba plugin of Cytoscape software coupled with analysis of enriched genes in the cell cycle pathway identified PTTG1 and CDC20 (two hub genes) as key targets associated with BPA-induced MCF-7 cell proliferation and migration. Interestingly, BPA significantly increased the protein expression levels of PTTG1 but not CDC20. Knockdown of PTTG1 inhibited the BPA-induced increase in proliferation and maintained cell cycle progression. In addition, we confirmed that the increased expression of PTTG1 upon BPA exposure was caused by miR-381-3p inhibition. Moreover, we verified that miR-381-3p expression was low and inversely correlated with PTTG1 expression in breast cancer tissues. Together, these findings demonstrate that BPA promotes high PTTG1 expression and alters the cell cycle to enhance MCF-7 cell proliferation by inhibiting miR-381-3p expression.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Compuestos de Bencidrilo , Neoplasias de la Mama/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Fenoles
17.
Toxicol In Vitro ; 62: 104686, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31614173

RESUMEN

Cadmium exposure has raised great public concern. Extensive studies have revealed the neurotoxic effects of cadmium exposure during brain development. However, more evidence is still needed to reach a consistent conclusion and uncover the underlying mechanisms. Here, we used primary mouse embryonic neural stem/progenitor cells (NSPCs) as a cell model and exposed the cells to 0, 1, 2 or 4 µM cadmium. High-throughput mRNA-seq technology was used to explore the global transcriptome changes in NSPCs after exposure to 2 µM cadmium. We found that cadmium exposure remarkably influenced the expression of genes involved in cell growth, proliferation, cell cycle and survival. Pathway-Act-Network analysis revealed that these altered genes were targeted to the P53, PI3K-AKT, MAPK, calcium, and NF-kappa B signaling pathways. In vitro experiments using cultured NSPCs verified that cadmium exposure reduced cell viability, proliferation, neurosphere formation and caused cell cycle arrest at low concentrations (≤ 2 µM), while induced cell apoptosis at high concentrations (≥ 4 µM). Real-time PCR results confirmed the concentration-dependent effects of cadmium exposure on the expression of critical genes in the above signaling pathways. Together, our results provide transcriptomic insight into cadmium-induced developmental neurotoxic effects and the underlying mechanisms.


Asunto(s)
Cadmio/toxicidad , Células-Madre Neurales/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Transcriptoma/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos BALB C , Neurogénesis/efectos de los fármacos , Embarazo , ARN/biosíntesis , ARN/genética , Transducción de Señal/efectos de los fármacos
18.
Toxicol Lett ; 246: 7-16, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-26809137

RESUMEN

Cadmium (Cd) is an important and common environmental pollutant that has been linked to cardiovascular diseases, such as atherosclerosis and hypertension. Increasing evidence demonstrates that Cd impairs the cardiovascular system by targeting vascular endothelial cells, but the underlying mechanisms remain obscure. In human umbilical vein endothelial cells (HUVECs), we observed that Cd treatment led to cell death and the generation of inflammatory cytokines. The Cd-induced cell death was identified as pyroptosis, a novel pro-inflammatory form of cell death depending on caspase-1 activation. In addition, exposure of HUVECs to Cd resulted in NLRP3 inflammasome activation as evidenced by cleavage of caspase-1 and downstream interleukin (IL)-1ß production. Moreover, knockdown of NLRP3 by small interfering RNA efficiently suppressed Cd-induced caspase-1 cleavage, IL-1ß production and pyroptosis in HUVECs. Additional experiments demonstrated that treatment with Cd significantly increased the levels of mitochondrial reactive oxygen species (mtROS) and intracellular ROS in HUVECs. Accordingly, pre-treatment with mtROS scavenger or total ROS scavenger reduced Cd-induced activation of NLRP3 inflammasome and pyroptotic cell death. Taken together, our data suggest that NLRP3 inflammasome, activated by the generation of mtROS, mediates Cd-induced pyroptosis in HUVECs. Our results provide novel insights into Cd-induced cytotoxicity and the underlying mechanism by which Cd induces endothelial injury.


Asunto(s)
Cadmio/farmacología , Proteínas Portadoras/fisiología , Células Endoteliales/efectos de los fármacos , Inflamasomas/fisiología , Piroptosis , Caspasa 1/fisiología , Células Cultivadas , Células Endoteliales/patología , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo
19.
Biomaterials ; 90: 27-39, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26986854

RESUMEN

Increased biomedical applications of quantum dots (QDs) have raised considerable concern regarding their toxicological impact. However, the toxicity of QDs is largely unknown and the underlying mechanism is still undefined. This study was conducted to examine the hepatotoxicity of CdSe/ZnS core/shell QDs and the underlying mechanism. In hepatic L02 cells, the QDs caused cytotoxicity in a dose-dependent manner. The QDs were then shown to activate the NLR pyrin domain containing 3 (NLRP3) inflammasome in hepatocytes, leading to a novel pro-inflammatory form of cell death named pyroptosis. Further experiments demonstrated that the QDs induced mitochondrial reactive oxygen species (mtROS) production, and that both a mtROS and a total ROS scavenger attenuated QDs-induced NLRP3 activation and pyroptosis. In addition, QDs increased cytoplasmic calcium (Ca(2+)) levels, while a Ca(2+) release antagonist and chelator alleviated QDs-induced mtROS, NLRP3 activation and subsequent pyroptosis in hepatocytes. In vivo, QDs administration induced liver inflammation and dysfunction. Moreover, the QDs also resulted in NLRP3 activation in liver tissue. However, QDs-induced liver inflammation and dysfunction were abolished in NLRP3 knockout mice. Also, an elevation in mtROS was observed in liver after QDs administration, and the mtROS scavenger suppressed liver NLRP3 activation, inflammation and dysfunction induced by QDs. Our data suggest that QDs induced hepatocyte pyroptosis, liver inflammation and dysfunction via NLRP3 activation, which was caused by QDs-triggered mtROS production and Ca(2+) mobilization. Our results provide novel insights into QDs-induced hepatotoxicity and the underlying mechanism, facilitating control of the side effects of QDs.


Asunto(s)
Compuestos de Cadmio/efectos adversos , Hepatocitos/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Piroptosis/efectos de los fármacos , Puntos Cuánticos/efectos adversos , Compuestos de Selenio/efectos adversos , Sulfuros/efectos adversos , Compuestos de Zinc/efectos adversos , Animales , Compuestos de Cadmio/química , Compuestos de Cadmio/inmunología , Línea Celular , Hepatocitos/inmunología , Humanos , Inflamasomas/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Hígado/efectos de los fármacos , Hígado/inmunología , Masculino , Ratones Endogámicos C57BL , Puntos Cuánticos/química , Especies Reactivas de Oxígeno/inmunología , Compuestos de Selenio/química , Compuestos de Selenio/inmunología , Sulfuros/inmunología , Compuestos de Zinc/inmunología
20.
PLoS One ; 11(3): e0150923, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26950212

RESUMEN

Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Campos Electromagnéticos , Células Madre Embrionarias/citología , Células-Madre Neurales/citología , Neuritas/efectos de la radiación , Canales Catiónicos TRPC/genética , Regulación hacia Arriba/efectos de la radiación , Animales , Encéfalo/embriología , Encéfalo/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Madre Embrionarias/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Células-Madre Neurales/efectos de la radiación , Neuritas/metabolismo , ARN Interferente Pequeño/genética , Canales Catiónicos TRPC/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA