Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 239(4): e31190, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219075

ABSTRACT

Selenium (Se), as one of the essential trace elements, plays an anti-inflammatory, antioxidation, and immune-enhancing effect in the body. In addition, Se can also improve nervous system damage induced by various factors. Earlier studies have described the important role of mitochondrial dynamic imbalance in lipopolysaccharide (LPS)-induced nerve injury. The inositol 1,4,5-triphosphate receptor (IP3R)/glucose-regulated protein 75 (GRP75)/voltage-dependent anion channel 1 (VDAC1) complex is considered to be the key to regulating mitochondrial dynamics. However, it is not clear whether Selenomethionine (SeMet) has any influence on the IP3R/GRP75/VDAC1 complex. Therefore, the aim of this investigation was to determine whether SeMet can alleviate LPS-induced brain damage and to elucidate the function of the IP3R/GRP75/VDAC1 complex in it. We established SeMet and/or LPS exposure models in vivo and in vitro using laying hens and primary chicken nerve cells. We noticed that SeMet reversed endoplasmic reticulum stress (ERS) and the imbalance in mitochondrial dynamics and significantly prevented the occurrence of neuronal apoptosis. We made this finding by morphological observation of the brain tissue of laying hens and the detection of related genes such as ERS, the IP3R/GRP75/VDAC1 complex, calcium signal (Ca2+), mitochondrial dynamics, and apoptosis. Other than that, we also discovered that the IP3R/GRP75/VDAC1 complex was crucial in controlling Ca2+ transport between the endoplasmic reticulum and the mitochondrion when SeMet functions as a neuroprotective agent. In summary, our results revealed the specific mechanism by which SeMet alleviated LPS-induced neuronal apoptosis for the first time. As a consequence, SeMet has great potential in the treatment and prevention of neurological illnesses (like neurodegenerative diseases).


Subject(s)
Apoptosis , HSP70 Heat-Shock Proteins , Membrane Proteins , Mitochondrial Dynamics , Neurons , Selenomethionine , Animals , Female , Apoptosis/drug effects , Calcium/metabolism , Chickens , Lipopolysaccharides/pharmacology , Selenomethionine/pharmacology , Voltage-Dependent Anion Channel 1/genetics , Neurons/drug effects
2.
Mol Ther ; 32(2): 540-555, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38213030

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific neutralizing antibodies (NAbs) lack cross-reactivity between SARS-CoV species and variants and fail to mediate long-term protection against infection. The maintained protection against severe disease and death by vaccination suggests a role for cross-reactive T cells. We generated vaccines containing sequences from the spike or receptor binding domain, the membrane and/or nucleoprotein that induced only T cells, or T cells and NAbs, to understand their individual roles. In three models with homologous or heterologous challenge, high levels of vaccine-induced SARS-CoV-2 NAbs protected against neither infection nor mild histological disease but conferred rapid viral control limiting the histological damage. With no or low levels of NAbs, vaccine-primed T cells, in mice mainly CD8+ T cells, partially controlled viral replication and promoted NAb recall responses. T cells failed to protect against histological damage, presumably because of viral spread and subsequent T cell-mediated killing. Neither vaccine- nor infection-induced NAbs seem to provide long-lasting protective immunity against SARS-CoV-2. Thus, a more realistic approach for universal SARS-CoV-2 vaccines should be to aim for broadly cross-reactive NAbs in combination with long-lasting highly cross-reactive T cells. Long-lived cross-reactive T cells are likely key to prevent severe disease and fatalities during current and future pandemics.


Subject(s)
Antibodies, Neutralizing , COVID-19 Vaccines , COVID-19 , Animals , Humans , Mice , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , Viral Vaccines
3.
Ecotoxicol Environ Saf ; 270: 115923, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38171107

ABSTRACT

3,3',4',4',5-Polychlorinated biphenyls (PCB126) is classified as a persistent organic environmental pollutant that can cause liver damage by producing excessive reactive oxygen species (ROS). ROS also can stimulate neutrophil extracellular traps (NETs) formation, which cause damage to organism if NETs are produced in excess. Melatonin is generally considered to possess strong antioxidant and anti-inflammation prosperities, but it is unclear whether it can alleviate PCB126-induced injury. To explore whether PCB126-induced liver injury is related to the formation of NETs and whether melatonin has a potent protective effect, we established PCB126 exposure/ PCB126 and melatonin co-treatment mouse models by gavage. To further clarify the specific mechanism, we also cultured neutrophils and AML12 cells to replicate in vivo model. Here, we found PCB126 exposure resulted in an elevation in the activities of MDA, LPO, PCO, and 8-OHdG, and a reduction in the activities of CAT, GSH-PX and SOD. We found that PCB126 exposure led to an elevation in the expression levels of chemokines (CCL2, CCL3, CCL4, CXCL12, and CXCL8) and marker factors for NETs formation (MPO, NE, NOX2, PKCα, and PKCζ) in the PCB126 group. IF, SYTOX staining, and SEM results also revealed that PCB126 could stimulate NETs formation. In addition, results of a co-culture system of PBNs and AML12 cells revealed that the expression levels of inflammatory cytokines (IL-1ß, IL-6, and TNF-α) significantly decreased and the expression levels of metabolism factors (Fas, Acc, and Srebp) slightly decreased for scavenging NETs, indicating NETs formation aggravated PCB126-induced hepatic damages. Noteworthy, treatment with melatonin reversed these results. In summary, our findings revealed that melatonin alleviated hepatic damage aggravated by PCB126-induced ROS-dependent NETs formation through suppressing excessive ROS production. This finding not only enriches toxicological mechanism of PCB126, but more importantly extends biological effects of melatonin and its potential application values.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Extracellular Traps , Melatonin , Polychlorinated Biphenyls , Mice , Animals , Extracellular Traps/metabolism , Polychlorinated Biphenyls/toxicity , Polychlorinated Biphenyls/metabolism , Reactive Oxygen Species/metabolism , Melatonin/pharmacology , Melatonin/metabolism , Lipid Metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Neutrophils/metabolism
4.
Cell Rep ; 42(8): 112961, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37561633

ABSTRACT

Refractory and relapsed B cell lymphomas are often driven by the difficult-to-target oncogene MYC. Here, we report that high MYC expression stimulates proliferation and protects B lymphoma cells from apoptosis under normal oxidative stress levels and that compounds including N-acetylcysteine (NAC) and vitamin C (VitC) induce apoptosis by reducing oxidative stress. NAC and VitC injections effectively reduce tumor growth in lymphoma cells with high MYC expression but not in those with low MYC expression. MYC knockdown confers tumor resistance to NAC and VitC, while MYC activation renders B cells sensitive to these compounds. Mechanistically, NAC and VitC stimulate MYC binding to EGR1 through Cys117 of MYC, shifting its transcriptional output from cell cycle to apoptosis gene expression. These results identify a redox-controlled mechanism for MYC's role in maintaining proliferation and preventing apoptosis, offering a potential therapeutic rationale for evaluating NAC or VitC in patients with MYC-driven B cell lymphoma.

5.
J Clin Invest ; 133(20)2023 10 16.
Article in English | MEDLINE | ID: mdl-37651203

ABSTRACT

Lung cancer progression relies on angiogenesis, which is a response to hypoxia typically coordinated by hypoxia-inducible transcription factors (HIFs), but growing evidence indicates that transcriptional programs beyond HIFs control tumor angiogenesis. Here, we show that the redox-sensitive transcription factor BTB and CNC homology 1 (BACH1) controls the transcription of a broad range of angiogenesis genes. BACH1 is stabilized by lowering ROS levels; consequently, angiogenesis gene expression in lung cancer cells, tumor organoids, and xenograft tumors increased substantially following administration of vitamins C and E and N-acetylcysteine in a BACH1-dependent fashion under normoxia. Moreover, angiogenesis gene expression increased in endogenous BACH1-overexpressing cells and decreased in BACH1-knockout cells in the absence of antioxidants. BACH1 levels also increased upon hypoxia and following administration of prolyl hydroxylase inhibitors in both HIF1A-knockout and WT cells. BACH1 was found to be a transcriptional target of HIF1α, but BACH1's ability to stimulate angiogenesis gene expression was HIF1α independent. Antioxidants increased tumor vascularity in vivo in a BACH1-dependent fashion, and overexpressing BACH1 rendered tumors sensitive to antiangiogenesis therapy. BACH1 expression in tumor sections from patients with lung cancer correlated with angiogenesis gene and protein expression. We conclude that BACH1 is an oxygen- and redox-sensitive angiogenesis transcription factor.


Subject(s)
Antioxidants , Basic-Leucine Zipper Transcription Factors , Lung Neoplasms , Humans , Antioxidants/pharmacology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Hypoxia , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Animals , Mice
6.
Redox Biol ; 60: 102619, 2023 04.
Article in English | MEDLINE | ID: mdl-36774779

ABSTRACT

Oxidative stress is a barrier of migration and metastasis for malignant melanoma cells. Consequently, reducing oxidative stress with the antioxidant N-acetylcysteine (NAC) stimulates melanoma cell migration in vitro and metastasis in vivo. However, it is not yet known whether the NAC effect is shared with other antioxidants. Here, we screened 104 redox-active compounds and identify 27 that increase migration of human malignant melanoma cells in two doses. Validation experiments in four cell lines and four drug doses resulted in a list of 18 compounds which were ranked based on their ability to increase migration and reduce ROS levels; vitamin C (VitC) ranked as number one, followed by the vitamin E analogue Trolox and several carotenoids and Vitamin A-related compounds. Four diet-relevant compounds from this list-VitC, ß-carotene, retinyl palmitate, and canthaxanthin-were selected and found to accelerate metastasis in mice with BRAFV600E-driven malignant melanoma. Genomics analyses revealed that the transcription factor BACH1 is activated following antioxidant administration and knockout of Bach1 in mouse melanoma cells reduced lymph node and liver metastasis in xenograft mouse models. We conclude that a broad range of antioxidants accelerate melanoma migration and metastasis and that BACH1 is functionally linked to melanoma metastasis in vivo.


Subject(s)
Antioxidants , Melanoma , Animals , Humans , Mice , Acetylcysteine , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Reactive Oxygen Species/metabolism , Vitamins , Vitamin A/pharmacology , Melanoma, Cutaneous Malignant
7.
Basic Clin Pharmacol Toxicol ; 131(4): 217-223, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35790078

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder characterized by premature ageing and early death at a mean age of 14.7 years. At the molecular level, HGPS is caused by a de novo heterozygous mutation in LMNA, the gene encoding A-type lamins (mainly lamin A and C) and nuclear proteins, which have important cellular functions related to structure of the nuclear envelope. The LMNA mutation leads to the synthesis of a truncated prelamin A protein (called progerin), which cannot undergo normal processing to mature lamin A. In normal cells, prelamin A processing involves four posttranslational processing steps catalysed by four different enzymes. In HGPS cells, progerin accumulates as a farnesylated and methylated intermediate in the nuclear envelope where it is toxic and causes nuclear shape abnormalities and senescence. Numerous efforts have been made to target and reduce the toxicity of progerin, eliminate its synthesis and enhance its degradation, but as of today, only the use of farnesyltransferase inhibitors is approved for clinical use in HGPS patients. Here, we review the main current strategies that are being evaluated for treating HGPS, and we focus on efforts to target the posttranslational processing of progerin.


Subject(s)
Progeria , Adolescent , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Farnesyltranstransferase/therapeutic use , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Nuclear Proteins/genetics , Progeria/drug therapy , Progeria/genetics , Progeria/metabolism , Protein Processing, Post-Translational
8.
Nat Commun ; 13(1): 3068, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35654881

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature ageing disorder caused by a point mutation in the LMNA gene (LMNA c.1824 C > T), resulting in the production of a detrimental protein called progerin. Adenine base editors recently emerged with a promising potential for HGPS gene therapy. However adeno-associated viral vector systems currently used in gene editing raise concerns, and the long-term effects of heterogeneous mutation correction in highly proliferative tissues like the skin are unknown. Here we use a non-integrative transient lentiviral vector system, expressing an adenine base editor to correct the HGPS mutation in the skin of HGPS mice. Transient adenine base editor expression corrected the mutation in 20.8-24.1% of the skin cells. Four weeks post delivery, the HGPS skin phenotype was improved and clusters of progerin-negative keratinocytes were detected, indicating that the mutation was corrected in both progenitor and differentiated skin cells. These results demonstrate that transient non-integrative viral vector mediated adenine base editor expression is a plausible approach for future gene-editing therapies.


Subject(s)
Progeria , Adenine , Animals , Lamin Type A/genetics , Lamin Type A/metabolism , Mice , Mutation , Phenotype , Progeria/genetics , Progeria/metabolism , Progeria/therapy
9.
Elife ; 102021 02 02.
Article in English | MEDLINE | ID: mdl-33526168

ABSTRACT

A farnesylated and methylated form of prelamin A called progerin causes Hutchinson-Gilford progeria syndrome (HGPS). Inhibiting progerin methylation by inactivating the isoprenylcysteine carboxylmethyltransferase (ICMT) gene stimulates proliferation of HGPS cells and improves survival of Zmpste24-deficient mice. However, we don't know whether Icmt inactivation improves phenotypes in an authentic HGPS mouse model. Moreover, it is unknown whether pharmacologic targeting of ICMT would be tolerated by cells and produce similar cellular effects as genetic inactivation. Here, we show that knockout of Icmt improves survival of HGPS mice and restores vascular smooth muscle cell numbers in the aorta. We also synthesized a potent ICMT inhibitor called C75 and found that it delays senescence and stimulates proliferation of late-passage HGPS cells and Zmpste24-deficient mouse fibroblasts. Importantly, C75 did not influence proliferation of wild-type human cells or Zmpste24-deficient mouse cells lacking Icmt, indicating drug specificity. These results raise hopes that ICMT inhibitors could be useful for treating children with HGPS.


Subject(s)
Cellular Senescence/drug effects , Progeria/drug therapy , Protein Methyltransferases/drug effects , Pyrans/pharmacology , Animals , Aorta/pathology , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Lamin Type A/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle , Progeria/genetics , Progeria/pathology , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism
10.
Aging Cell ; 19(8): e13200, 2020 08.
Article in English | MEDLINE | ID: mdl-32910507

ABSTRACT

Several progeroid disorders are caused by deficiency in the endoprotease ZMPSTE24 which leads to accumulation of prelamin A at the nuclear envelope. ZMPSTE24 cleaves prelamin A twice: at the third carboxyl-terminal amino acid following farnesylation of a -CSIM motif; and 15 residues upstream to produce mature lamin A. The carboxyl-terminal cleavage can also be performed by RAS-converting enzyme 1 (RCE1) but little is known about the importance of this cleavage for the ability of prelamin A to cause disease. Here, we found that knockout of RCE1 delayed senescence and increased proliferation of ZMPSTE24-deficient fibroblasts from a patient with non-classical Hutchinson-Gilford progeria syndrome (HGPS), but did not influence proliferation of classical LMNA-mutant HGPS cells. Knockout of Rce1 in Zmpste24-deficient mice at postnatal week 4-5 increased body weight and doubled the median survival time. The absence of Rce1 in Zmpste24-deficient fibroblasts did not influence nuclear shape but reduced an interaction between prelamin A and AKT which activated AKT-mTOR signaling and was required for the increased proliferation. Prelamin A levels increased in Rce1-deficient cells due to a slower turnover rate but its localization at the nuclear rim was unaffected. These results strengthen the idea that the presence of misshapen nuclei does not prevent phenotype improvement and suggest that targeting RCE1 might be useful for treating the rare progeroid disorders associated with ZMPSTE24 deficiency.


Subject(s)
Genes, ras/genetics , Membrane Proteins/deficiency , Metalloendopeptidases/deficiency , Progeria/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Phenotype
11.
Food Funct ; 11(2): 1312-1321, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32022057

ABSTRACT

Over the past decades, substantial advances have been made in both the early diagnosis and accurate prognosis of numerous cancers because of the impressive development of novel proteomic strategies. Selenium (Se) is an essential trace element in humans and animals. Se deficiency could lead to Keshan disease in humans, mulberry heart disease in pigs and damage of tissues including cardiac injury, apoptosis in the liver, reduction in the immune responses in spleen and cerebral lesions in chickens. However, it is well know that plasma biomarkers are not specific and also show alterations in various diseases including those caused by Se deficiency. Therefore, new definition biomarkers are needed to improve disease surveillance and reduce unnecessary chicken losses due to Se deficiency. To identify new biomarkers for Se deficiency, we performed exploratory heart, liver, spleen, muscle, vein, and artery proteomic screens to further validate the biomarkers using Venn analysis, GO enrichment, heatmap analysis, and IPA analysis. Based on the bioinformatics methods mentioned above, we found that differentially expressed genes and proteins are enriched to the PI3K/AKT/mTOR signal pathway and insulin pathway. We further used western blot to detect the expression of proteins related to the two pathways. Results showed that the components of the PI3K/AKT/mTOR signal pathway were definitely decreased in heart, liver, spleen, muscle, vein and artery tissues in the Se deficient group. Expression IGF and IGFBP2 of the insulin pathway were differentially increased in the heart, liver, and spleen in Se deficient group samples and decreased in muscle and artery. In conclusion, 5 proteins, namely PI3K, AKT, mTOR, IGF, and IGFBP2, were differentially expressed, which could be potentially useful Se deficient biomarkers. In the present study, proteomic profiling was used to elucidate protein biomarkers that distinguished Se deficient samples from the controls, which might provide a new direction for the diagnosis and targeted treatment induced by Se deficiency in chickens.


Subject(s)
Organ Specificity/physiology , Proteome , Selenium , Signal Transduction/physiology , Animals , Apoptosis/physiology , Biomarkers , Chickens , Proteome/analysis , Proteome/chemistry , Proteome/metabolism , Proteomics , Selenium/deficiency , Selenium/metabolism
12.
Cell ; 178(2): 330-345.e22, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31257027

ABSTRACT

For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.


Subject(s)
Antioxidants/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Glycolysis/drug effects , Lung Neoplasms/pathology , Animals , Antioxidants/administration & dosage , Basic-Leucine Zipper Transcription Factors/genetics , Cell Movement/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Heme/metabolism , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Hexokinase/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
13.
Metallomics ; 10(5): 759-767, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29766201

ABSTRACT

Selenium (Se) deficiency inhibits immune cell differentiation, affects immune response, and leads to cellular and humoral immune dysfunction. However, the impact of Se deficiency on the differentiation and Th1/Th2 balance of dendritic cells is still unclear. In this study, we replicated a model of Se-deficient chickens by feeding the chickens with a low-Se diet (i.e., the content of Se is 0.008 mg per kg diet). On this basis, we explored the effect of Se deficiency on the differentiation of chicken dendritic cells by induction culture of peripheral blood monocyte cells. We induced chicken dendritic cells by incubating mononuclear cells with a 100 ng mL-1 recombinant chicken granulocyte-macrophage colony-stimulating factor and 20 ng mL-1 recombinant chicken IL-4 for total 7 days. The results showed that Se deficiency decreased the expression of cell-surface markers including CD11c, CD40, CD86, and MHC II. Furthermore, we analyzed the cytokine profiles using real-time quantitative PCR and ELISA. The results indicated that Se deficiency inhibited the expression of selenoproteins and changed the secretion of IL-10, IL-12p40, and IFN-γ. Additionally, Se deficiency weakened the ability of dendritic cells to stimulate the proliferation of mixed allogeneic lymphocytes. In conclusion, Se deficiency suppressed the differentiation and immune function of chicken dendritic cells by down-regulating the expression of CD11c, CD40, CD86, MHC II, and selenoproteins. The result also showed that the Th1/Th2 imbalance was induced by enhancing the secretion of Th1-type cytokine IL-12p40 and IFN-γ and reducing that of Th2-type cytokine IL-10. Our findings contribute to understanding the mechanism of Se deficiency in the differentiation and immune function of chicken dendritic cells.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Monocytes/immunology , Selenium/deficiency , Selenoproteins/metabolism , Th1-Th2 Balance , Animals , Cells, Cultured , Chickens , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation , Selenoproteins/genetics
14.
PLoS One ; 12(9): e0184186, 2017.
Article in English | MEDLINE | ID: mdl-28877212

ABSTRACT

Ion homeostasis plays important roles in development of metabolic diseases. In the present study, we examined the contents and distributions of 25 ions in chicken muscles following treatment with selenium (Se) deficiency for 25 days. The results revealed that in chicken muscles, the top ranked microelements were silicon (Si), iron (Fe), zinc (Zn), aluminum (Al), copper (Cu) and boron (B), showing low contents that varied from 292.89 ppb to 100.27 ppm. After Se deficiency treatment, essential microelements [Cu, chromium (Cr), vanadium (V) and manganese (Mn)], and toxic microelements [cadmium (Cd) and mercury (Hg)] became more concentrated (P < 0.05). Elements distribution images showed generalized accumulation of barium (Ba), cobalt (Co), Cu, Fe and V, while Cr, Mn, and Zn showed pin point accumulations in muscle sections. Thus, the ion profiles were generally influenced by Se deficiency, which suggested a possible role of Se deficiency in muscle dysfunctions caused by these altered ion profiles.


Subject(s)
Muscle, Skeletal/metabolism , Selenium/deficiency , Trace Elements/metabolism , Aluminum/analysis , Aluminum/metabolism , Animals , Boron/analysis , Boron/metabolism , Chickens , Chromium/analysis , Chromium/metabolism , Copper/analysis , Copper/metabolism , Ions/analysis , Ions/metabolism , Iron/analysis , Iron/metabolism , Male , Manganese/analysis , Manganese/metabolism , Muscle, Skeletal/chemistry , Silicon/analysis , Silicon/metabolism , Trace Elements/analysis , Vanadium/analysis , Vanadium/metabolism , Zinc/analysis , Zinc/metabolism
15.
Ecotoxicol Environ Saf ; 145: 503-510, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28783600

ABSTRACT

The nuclear transcription factor NF-E2-related factor 2 (Nrf2) binds to antioxidant response elements (AREs) and is involved in the regulation of genes participated in defending cells against oxidative damage, which have been confirmed in animal models. Selenium (Se), known as an important element in the regulation of antioxidant activity, can antagonize Cadmium (Cd) toxicity in birds. However, the role of Nrf2 in selenium-cadmium interaction has not been reported in birds. To further explore the mechanism of selenium attenuating spleen toxicity induced by cadmium in chickens, cadmium chloride (CdCl2, 150mg/kg) and sodium selenite (Na2SeO3, 2mg/kg) were co-administrated or individually administered in the diet of chickens for 90 days. The results showed that Cd exposure increased the level of hydrogen peroxide (H2O2) and malondialdehyde (MDA) and decreased the antioxidant enzyme activities, including superoxide dismutase (SOD), glutathione peroxidase (Gpx), total antioxidative capacity (T-AOC), catalase (CAT). Cd exposure increased obviously nuclear accumulation of Nrf2, and the expression of Nrf2 downstream heme oxygenase-1 (HO-1) and NAD(P)H: quinine oxidoreductase 1 (NQO1), reduced the expression of Kelch-like ECH-associated protein (keap1), Gpx-1 and thioredoxin reductase-1 (TrxR1). In addition, Cd induced the increase of bak, caspase9, p53, Cyt c mRNA levels, increased bax/bcl-2 ratio, increased caspase3 mRNA and protein levels. Selenium treatment reduced the accumulation of Cd in the spleen, attenuates Cd-induced Nrf2 nuclear accumulation, enhanced antioxidant enzyme activities, ameliorated Cd-induced oxidative stress and apoptosis in the spleen. In summary, our results demonstrate that Se ameliorated spleen toxicity induced by cadmium by modulating the antioxidant system, independently of Nrf2-regulated antioxidant response pathway.


Subject(s)
Antioxidants/metabolism , Apoptosis/drug effects , Cadmium/toxicity , Chickens/metabolism , Environmental Pollutants/toxicity , NF-E2-Related Factor 2/metabolism , Selenium/pharmacology , Spleen/drug effects , Animals , Catalase/metabolism , Dose-Response Relationship, Drug , Oxidative Stress/drug effects , Selenium/metabolism , Spleen/metabolism , Spleen/pathology , Superoxide Dismutase/metabolism
16.
Dev Comp Immunol ; 77: 30-37, 2017 12.
Article in English | MEDLINE | ID: mdl-28735963

ABSTRACT

Selenium (Se) promotes immune cell differentiation and improves immune response. Antigen-presenting cells such as dendritic cells (DCs) play an important role in immune system, however, the impact of Se on DCs is still unclear. In this study, we successfully induced and cultured chicken DCs from peripheral blood mononuclear cells by incubating mononuclear cells with 50 ng/mL recombinant chicken granulocyte-macrophage colony stimulating factor and 10 ng/mL recombinant chicken interleukin-4 for total 9 days. In + Se group, we added 10-7 mol/L sodium selenite from the first day of cell culture. The results showed that Se supplementation expedited and increased the expression of cell surface markers including CD11c, CD40, CD86, and MHC II. Principal component analysis showed that the expression of selenoproteins SelW, SelK, Dio3, GPX1, GPX2, SelN, SelS, SelH in chicken DCs was highly correlated, and SelW had highest correlation with the cell surface markers MHC II and CD11c. In conclusion, Se accelerates the differentiation and maturation of chicken DCs. Se regulates the differentiation and maturation of chicken DCs by selenoproteins. Selenoproteins has closely correlated to surface markers of chicken DCs.


Subject(s)
Dendritic Cells/immunology , Selenium/metabolism , Selenoproteins/metabolism , Animals , Antigen Presentation , Antigens, CD/metabolism , Cell Differentiation , Cells, Cultured , Chickens , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Histocompatibility Antigens Class II/metabolism , Interleukin-4/metabolism , Selenoproteins/genetics , Sodium Selenite/metabolism
17.
Biol Trace Elem Res ; 180(1): 135-145, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28281222

ABSTRACT

In the present study, specific small interfering RNA (siRNA) for selenoprotein K (Selk) gene was designed and transfected into chicken myoblasts. Then, the expressions of inflammatory factors (including induced nitric oxide synthase [iNOS], nuclear factor-kappa B [NF-κB], heme-oxygenase-1 [HO-1], cyclooxygenase-2 [COX-2], and prostaglandin E synthase [PTGEs]), inflammation-related cytokines (including interleukin [IL]-1ß, IL-6, IL-7, IL-8, IL-17, and interferon [IFN]-γ), and heat shock proteins (HSPs) (including HSP27, HSP40, HSP60, HSP70, and HSP90) were examined at 24 and 72 h after transfection. The results showed that messenger RNA (mRNA) expressions of iNOS, NF-κB, HO-1, COX-2, IL-6, IL-7, IL-8, HSP 27, HSP 40, HSP 60, HSP 70, and HSP 90 were significantly increased (p < 0.05) at 24 and 72 h after siRNA transfection, and the mRNA expressions of PTGEs, IL-1ß, IL-17, and IFN-γ were significantly increased and decreased (p < 0.05) at 24 and 72 h after siRNA transfection. The results also showed that the protein expressions of iNOS, NF-κB, HO-1, COX-2, HSP60, HSP70, and HSP90 were significantly increased (p < 0.05) at 24 and 72 h after siRNA transfection. The correlation analysis and principal component analysis (PCA) showed that PTGEs, IL-1ß, IL-17, IFN-γ, HSP40, and HSP90 might play special roles in response to Selk silencing in chicken myoblasts. These results indicated that Selk silencing induced inflammation response by affecting the expression levels of inflammatory factors and inflammation-related cytokines, and the heat shock proteins might play protective roles in this response in chicken myoblasts.


Subject(s)
Heat-Shock Proteins/genetics , Inflammation/genetics , Myoblasts/physiology , Selenoproteins/genetics , Animals , Chick Embryo , Cyclooxygenase 2/genetics , Cytokines/genetics , Gene Expression Regulation , Gene Silencing , Heme Oxygenase-1/genetics , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Principal Component Analysis , RNA, Small Interfering , Selenoproteins/metabolism
18.
J Vet Sci ; 18(3): 333-340, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-27586463

ABSTRACT

The aim of this study was to evaluate the influence of phosphorus (P) deficiency on the morphological and functional characteristics of erythrocytes in cows. Forty Holstein-Friesian dairy cows in mid-lactation were randomly divided into two groups of 20 each and were fed either a low-P diet (0.03% P/kg dry matter [DM]) or a control diet (0.36% P/kg DM). Red blood cell (RBC) indices results showed RBC and mean corpuscular hemoglobin decreased while mean corpuscular volume increased significantly (p < 0.05) in P-deficient cows. Erythrocyte morphology showed erythrocyte destruction in P-deficient cows. Erythrocytes' functional characteristics results showed total bilirubin and indirect bilirubin concentrations and aspartate transaminase and alanine transaminase activity levels in the serum of P-deficient cows were significantly higher than those in control diet-fed cows. Activities of superoxide dismutase and glutathione peroxidase in erythrocytes were lower, while the malondialdehyde content was greater, in P-deficient cows than in control diet-fed cows. Na+/K+-ATPase and Mg2+-ATPase activities were lower in P-deficient cows than in control diet-fed cows; however, Ca2+-ATPase activity was not significantly different. The phospholipid composition of the erythrocyte membrane changed and membrane fluidity rigidified in P-deficient cows. The results indicate that P deficiency might impair erythrocyte integrity and functional characteristics in cows.


Subject(s)
Erythrocytes/pathology , Phosphorus/deficiency , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Cattle/blood , Erythrocyte Indices , Erythrocytes/physiology , Female , Glutathione Peroxidase/blood , Sodium-Potassium-Exchanging ATPase/blood , Superoxide Dismutase/blood
19.
Oncotarget ; 7(36): 57618-57632, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27557522

ABSTRACT

Selenium (Se) deficiency induces Ca2+ leak and calcification in mammal skeletal muscles; however, the exact mechanism is still unclear. In the present study, both Se-deficient chicken muscle models and selenoprotein W (SelW) gene knockdown myoblast and embryo models were used to study the mechanism. The results showed that Se deficiency-induced typical muscular injuries accompanied with Ca2+ leak and oxidative stress (P < 0.05) injured the ultrastructure of the sarcoplasmic reticulum (SR) and mitochondria; decreased the levels of the Ca2+ channels, SERCA, SLC8A, CACNA1S, ORAI1, STIM1, TRPC1, and TRPC3 (P < 0.05); and increased the levels of Ca2+ channel PMCA (P < 0.05). Similarly, SelW knockdown also induced Ca2+ leak from the SR and cytoplasm; increased mitochondrial Ca2+ levels and oxidative stress; injured SR and mitochondrial ultrastructure; decreased levels of SLC8A, CACNA1S, ORA1, TRPC1, and TRPC3; and caused abnormal activities of Ca2+ channels in response to inhibitors in myoblasts and chicken embryos. Thus, both Se deficiency and SelW knockdown induced Ca2+ leak, oxidative stress, and Ca2+ channel reduction. In addition, Ca2+ levels and the expression of the Ca2+ channels, RyR1, SERCA, CACNA1S, TRPC1, and TRPC3 were recovered to normal levels by N-acetyl-L-cysteine (NAC) treatment compared with SelW knockdown cells. Thus, with regard to the decreased Ca2+ channels, SelW knockdown closely correlated Se deficiency with Ca2+ leak in muscles. The redox regulation role of SelW is crucial in Se deficiency-induced Ca2+ leak in muscles.


Subject(s)
Calcium Channels/chemistry , Calcium/chemistry , Oxidation-Reduction , Selenium/deficiency , Selenoprotein W/chemistry , Acetylcysteine/chemistry , Animals , Antioxidants/chemistry , Calcinosis , Calcium/metabolism , Chick Embryo , Chickens , Cytosol/metabolism , Male , Membrane Potentials , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Oxidative Stress , Sarcoplasmic Reticulum/metabolism
20.
Biometals ; 29(4): 679-89, 2016 08.
Article in English | MEDLINE | ID: mdl-27344320

ABSTRACT

The aim of the present study was to clarify the effect of Selenoprotein K (Selk) silencing on the mRNA expression of 25 selenoproteins in chicken myoblasts. The specific small interfering RNA (siRNA) for Selk gene was designed and transfected into chicken myoblasts. Post-transfection mRNA expression of 25 selenoproteins was determined at various time periods i.e., 24, 48 and 72 h. Moreover, based on the results of expression of 25 selenoproteins, correlation analysis and principal component analysis (PCA) were used for further analysis. The results showed that the designed siRNA effectively inhibited Selk expression (decreased by 20, 29 and 43 % on 24, 48 and 72 h, respectively) and the mRNA expression levels of the 23 selenoproteins were influenced by silencing Selk differently (P < 0.05). Time-dependent pattern of mRNA expression after siRNA treatment in three groups were found similar: one group including Gpx1, Gpx2, Gpx3, Gpx4, Txnrd1, Txnrd2, Txnrd3, Sepw1, Selh, Sepp1, Selo and Sepx1, another group including Sepn1, Sels, Selt, Selm and Sep15 and other group including Dio2 and Dio3. The results of correlation analysis showed that Gpx1, Gpx2, Gpx3, Gpx4, Dio1, Dio3, Sepn1, Sels, Sepw1, Selt, Selh, Sep15, Seli and Selu had a positive correlation with Selk, while Dio2 and Sepp1 had a negative correlation with Selk. PCA data also indicated that Txnrd1, Txnrd2, Dio2, Selpb, Sepp1and Selo may play special roles in response to Selk silencing. In summary, these results indicated that different selenoproteins possess and exhibits distinct responses to silencing of Selk in chicken myoblasts.


Subject(s)
Gene Silencing , Myoblasts/metabolism , Selenoproteins/genetics , Animals , Cells, Cultured , Chickens , Gene Expression Profiling , Gene Silencing/drug effects , Myoblasts/cytology , Principal Component Analysis , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Selenoproteins/antagonists & inhibitors , Selenoproteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...