Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
2.
Biol Trace Elem Res ; 201(1): 368-376, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35080709

ABSTRACT

The aim of this study was to investigate the role of selenoproteins in Macranthoidin B (MB) with regard to the inhibition of hepa1-6 cell proliferation. The CCK8 method was used to detect the inhibition rate in hepa1-6 cell of proliferation. The production of ROS, MDA, GSH levels, and GSH-Px and SOD activities was detected according to corresponding reagent kits. We determined the mRNA expressions of 25 selenoproteins in hepa1-6 cells via real-time quantitative PCR (qRT-PCR); moreover, the heat map and principal component analysis were used for further bioinformatics analysis. The results revealed that with an increasing concentration of MB, the inhibitory effect on hepa1-6 cell proliferation intensified. Compared with the control group, the treatment group showed significantly increased ROS levels, elevated MDA contents, and decreased GSH level, GSH-Px activity, and SOD activity. Increasing MB concentration treatment induced remarkable degradation of Txnrd1, Txnrd2, Txnrd3, Gpx1, Gpx2, Gpx3, Gpx6, Dio1, Dio2, Selt, Selp, Selh, Selk, Selw, Seln, and Dio3. Principal component analysis revealed that Txnrd 3, Selk, Selo, Selw, Selt, Dio2, Txnrd1, Dio3, Gpx6, and Dio1 were highly correlated with MB. In conclusion, MB dose dependently inhibited hepa1-6 cell proliferation and induced oxidative stress. Based on bioinformatics analysis, with MB treatment, Txnrd 3, Selk, Selo, Selw, Selt, Dio2, Txnrd1, Dio3, Gpx6, and Dio1 exhibited critical role in the inhibition of hepa1-6 cells proliferation. The functions of these selenoproteins were associated with oxidative stress.


Subject(s)
Selenium , Mice , Animals , Reactive Oxygen Species , Selenium/pharmacology , Mice, Inbred Strains , Selenoproteins/metabolism , Oxidative Stress , Cell Proliferation , Superoxide Dismutase/metabolism
3.
Biofactors ; 47(5): 788-800, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34128579

ABSTRACT

Selenium (Se) plays a crucial role in intestinal health. However, the specific mechanism by which deficiency of Se causes intestinal damage remains unclear. This study was to explore whether Se deficiency can cause ER stress and induce apoptosis in swine small intestine. We established the Se deficiency swine model in vivo and the intestinal epithelial (IPEC-J2) cell Se deficiency model in vitro. The results of morphological observation showed that Se deficiency caused structural damage in intestinal villi and the decrease of goblet cell structure. The apoptotic characteristics such as nucleolar condensation, mitochondrial swelling, and apoptotic bodies were observed in the IPEC-J2 cells. The results of acridine orange/ethidium bromide and mitochondrial membrane potential fluorescence staining in vitro showed that there were more apoptotic cells in the Se-deficiency group than that in the control group. The protein and/or mRNA expression levels of Bax, Bcl-2, caspase 3, caspase 8, caspase 9, cytc, PERK, ATF6, IRE, XBP1, CHOP, GRP78, which are related to ER stress-apoptosis pathway, were significantly increased in the Se-deficient group which compared with the control group in vivo and in vitro were consistent. These results indicated that Se deficiency induced ER stress and increased the apoptosis in swine small intestine and IPEC-J2 cells and then caused the damage in swine small intestinal tissue. Besides, the results of gene expressions in our experiment proved that ER stress induced by Se deficiency promoted apoptosis. These results filled the blank in the mechanism of Se deficiency-induced intestinal injury in swine.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Intestine, Small/physiopathology , Selenium/deficiency , Animals , Disease Models, Animal , Swine
4.
J Cell Physiol ; 236(1): 157-177, 2021 01.
Article in English | MEDLINE | ID: mdl-32542694

ABSTRACT

Long noncoding RNAs (LncRNAs) have been demonstrated to be associated with a variety of myocardial diseases, but how LncRNAs regulate autophagy in selenium (Se)-deficient myocardial injury is infrequently reported. Here, we screened out a novel long noncoding RNA, microRNA, and ATG7 through transcriptomic results. We employed a Se-deficient chicken model in vivo, and primary cultured cardiomyocytes treated by correlation in vitro. The results showed that Se deficiency upregulated the expression of ATG7, and miR-17-5p inhibited cardiomyocyte autophagy by targeting ATG7. Furthermore, we found that LncRNA 0003250 regulated miR-17-5p, and thus affected the expression of ATG7 and autophagic cell death. Our present study proposed a novel model for the regulation of cardiomyocytes autophagy, which includes LncRNA 0003250, miR-17-5p and ATG7 in the chicken heart. Our conclusions may provide a feasible diagnostic tool for Se-deficient cardiomyocyte injury.


Subject(s)
Autophagy/genetics , Chickens/genetics , Heart/physiopathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Selenium/deficiency , Animals , Myocytes, Cardiac/pathology , Transcriptome/genetics , Up-Regulation/genetics
5.
Biol Trace Elem Res ; 194(1): 237-243, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31218646

ABSTRACT

Although it has been reported that selenium (Se) deficiency can trigger inflammation, however, there are few reports on the effect of Se on the function of mouse peritoneal macrophages. Herein, we examined the expression of inflammatory factors, oxidative stress levels, and phagocytosis for primary-cultured peritoneal macrophages using control and Se-deficient groups. Our results revealed that Se deficiency induced the accumulation of oxygen free radicals and weakened antioxidant capacity. Se deficiency also significantly increased the expression of inflammation factors including iNOS, IL-1ß, IL-12, IL-10, PTGe, and NF-κB. Meanwhile, Se suppression restrained macrophage production of TNF-α. The results of the phagocytosis assay demonstrated that Se deficiency inhibited the phagocytosis of macrophages. In conclusion, Se-deficient macrophages undergo severe inflammation through the NF-κB pathway due to the accumulation of oxygen free radicals and are hindered in their phagocytic capacity.


Subject(s)
Inflammation/drug therapy , Macrophages/drug effects , Phagocytosis/drug effects , Selenium/pharmacology , Animals , Cells, Cultured , Cytokines/analysis , Cytokines/genetics , Cytokines/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice , Oxidative Stress/drug effects , Selenium/deficiency
6.
J Inorg Biochem ; 193: 60-69, 2019 04.
Article in English | MEDLINE | ID: mdl-30684759

ABSTRACT

Our previous study revealed that selenium (Se) deficiency can cause myocardial injury through triggering autophagy. MicroRNAs (miRNAs) play crucial roles in autophagic cell death. However, the relationship between miRNAs and myocardial autophagy injury caused by Se deficiency remains unclear. We selected differential microRNA-215-5p (miR-215-5p) in Se-deficient myocardial tissue using high-throughput miRNA-sequencing. To further explore the role of miR-215-5p in myocardial injury, overexpression/knockdown of miR-215-5p in primary cardiomyocyte model was established by miRNAs interference technology. In this study, we report that miR-215-5p can promote myocardial autophagy by directly binding to the 3'untranslated region (3'UTR) of phosphatidylinositol-4, 5-bisphosphate 3-kinase (PI3K). Its target gene PI3K was confirmed by dual luciferase reporter assay, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot in cardiomyocytes. Our results showed that overexpression of miR-215-5p could trigger myocardial autophagy through PI3K-threonine-protein kinase (AKT)-target of rapamycin (TOR) pathway. Further studies revealed that autophagic cell death was dependent on the activation of extracellular signal-regulated kinase1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), p38 kinase (p38) and generation of reactive oxygen species (ROS) in overexpression of miR-215-5p in cardiomyocytes. On the contrary, miR-215-5p inhibitor can enhance cell survival capacity against autophagy by inhibiting ROS-mitogen-activated protein kinase (MAPK) pathways and activating the PI3K/AKT/TOR pathway in cardiomyocytes. Together, our findings support that miR-215-5p may modulate cell survival programs by regulating autophagy, and miR-215-5p acts as an autophagic regulator in the regulatory feedback loop that regulates cardiomyocyte survival by modulating the PI3K/AKT/TOR pathway and ROS-dependent MAPK pathways.


Subject(s)
Autophagy/physiology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , MAP Kinase Signaling System/physiology , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Animals , Cell Survival/physiology , Chickens , Class I Phosphatidylinositol 3-Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/cytology , Reactive Oxygen Species/metabolism , Selenium/deficiency
7.
Metallomics ; 11(2): 415-428, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30565618

ABSTRACT

Selenium (Se) is imperative for normal myocardial differentiation and development, and these basic cellular functions can be regulated by miRNA during cardiogenesis. Here, we show that Se deficiency can cause defects in myocardial development and abnormalities of cardiomyocyte differentiation. In previous work using microRNAome analysis, we found that miR-215-5p was differentially expressed in Se-deficient myocardial tissues. However, the relationship between miR-215-5p and Se deficiency in myocardial development remains unknown. In this study, CCCTC-binding factor (CTCF) was confirmed as the target gene of miR-215-5p by dual luciferase reporter assay, western blot and quantitative real-time polymerase chain reaction (qRT-PCR) in cardiomyocytes. Based on in vivo and in vitro results, we found that the increased expression of miR-215-5p induced by Se deficiency may cause transcriptional disorders of myocardial genes, mitochondrial biosynthesis imbalance, and a reduction of myocardial development and differentiation-related factors. Moreover, miR-215-5p may target CTCF to regulate myocardial development and differentiation via the noncanonical Wnt signaling pathway and induce mitochondrial dysfunction via the PGC-1α-TFAM-NRF1/2 pathway in the heart. Our results not only demonstrated that Se deficiency affected myocardial development and differentiation by directly targeting the miR-215-5p/CTCF axis but also found that miR-215-5p inhibitor promoted normal differentiation of cardiomyocytes and myocardial development and ameliorated myocardium structural abnormalities via the noncanonical Wnt signaling pathway in chicken. Our findings support the potential of applying miRNAs during the process of cardiogenesis and indicate that miR-215-5p could be a novel candidate for treatment of cardiac hypoevolutism.


Subject(s)
Myocytes, Cardiac/metabolism , Selenium/metabolism , 3' Untranslated Regions/genetics , 3' Untranslated Regions/physiology , Animals , Blotting, Western , Cell Differentiation/physiology , Chickens , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Selenium/deficiency , Software
8.
Cell Physiol Biochem ; 51(2): 778-792, 2018.
Article in English | MEDLINE | ID: mdl-30463056

ABSTRACT

BACKGROUND/AIMS: Selenium (Se) deficiency can lead to several cardiac diseases, including Keshan disease in humans, mulberry heart disease in pigs and cardiac injury in chickens. MicroRNAs have been a research focus in recent years and have been shown to participate in a new avenue of cell death-autophagy, which can play a significant role in several types of heart disease. METHODS: MicroRNAome analysis showed that the expression of miR-2954 was increased in the myocardium of selenium-deficient chickens, and PI3K was predicted to be the target gene. The target relationship between miR-2954 and PI3K was verified with a double fluorescence enzyme assay and RNA Protein Interaction Prediction and molecular docking software. qRT-PCR and western blotting were used to detect the expression of PI3K and related pathway components in selenium-deficient chickens and miR-2954 knockout/overexpression cardiomyocytes. RESULTS: In this study, we observed that miR-2954 overexpression led to inhibition of PI3K pathway in vivo and in vitroled to inhibition of the PI3K pathway in vivo and in vitro. CONCLUSION: The expression of miR-2954 was increased in selenium-deficient myocardium, whereas overexpression of miR-2954 led to autophagy and apoptosis of myocardial cells during cardiac injury through regulation of the PI3K pathway; whether this phenomenon is a self-protection mechanism of the organism or damage caused by miR-2954 requires further study. Our findings provides new insight apoptosis in cardiomyocytes; additionally, we aim to provide a new direction for the diagnosis and targeted treatment of myocardial diseases.


Subject(s)
Apoptosis , Autophagy , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Selenium/deficiency , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Autophagosomes/metabolism , Autophagosomes/pathology , Binding Sites , Caspase 3/genetics , Caspase 3/metabolism , Chickens , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Docking Simulation , Myocardium/metabolism , Myocardium/ultrastructure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nucleic Acid Conformation , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Transcriptome
9.
Biol Trace Elem Res ; 185(1): 162-169, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29264824

ABSTRACT

Lead (Pb) is a ubiquitous and toxic heavy metal and it can damage the immune system in humans and animals. Many researchers have reported that Selenium (Se) could possess various pharmacological effects in mammals. However, few studies have been carried out to investigate the protective role of Se in birds, especially in chickens. In this study, we investigated the protective effects of Se against Pb-induced inflammatory responses and the expression of heat shock proteins (HSPs) in peripheral blood neutrophils. One hundred eighty Hy-Line brown chickens were randomly divided into the control group (Con group), Se supplementation group (+Se group), Pb supplementation group (+Pb group), and the Se and Pb compound group (Se+Pb group). On the 90th day of the experiment, the peripheral blood was collected to extract neutrophils, and then, the levels of HSPs and cytokines were examined. The results showed that, after Pb treatment, the levels of IL-(1ß, 1R, 4, 8, 10, and 12ß), TGF-ß4, and HSP (27, 40, 60, 70, and 90) mRNA were significantly increased and levels of IL-2 and IFN-γ mRNA were decreased compared with those in the control group. Compared with the control group, the protein levels of HSP60 and HSP70 were also increased in the Pb treatment group. Co-administration of Se (1 mg/kg/day) and Pb resulted in a reversal of the Pb-induced cytokine changes in neutrophils accompanied by a significant decrease in HSPs. Our study demonstrated that Pb could decrease the immune function via changing the expression of cytokines and HSPs in chicken neutrophils, but Se could relieve the toxic effect induced by Pb.


Subject(s)
Heat-Shock Proteins/metabolism , Lead/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Selenium/pharmacology , Animals , Chaperonin 60/genetics , Chickens , HSP70 Heat-Shock Proteins/genetics , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-12/genetics , Interleukin-1beta/genetics , Interleukin-2/genetics , Interleukin-4/genetics , Interleukin-8/genetics
10.
J Inorg Biochem ; 177: 118-126, 2017 12.
Article in English | MEDLINE | ID: mdl-28957736

ABSTRACT

Thioredoxin (Txn) system is the most crucial antioxidant defense mechanism in myocardium. The aim of this study was to clarify the effect of Txn low expression on 25 selenoproteins in chicken cardiomyocytes. We developed a Se-deficient model (0.033mg/kg) and Txn knock down cardiomyocytes model (siRNA) studies. Western Blot, Quantitative Real-time PCR (qPCR) were performed, and correlation analysis, heat map were used for further analysis. Both low expression of Txn models are significantly decreased (P<0.05) the mRNA levels of Deiodinase 1, 2 (Dio 1, 2), Glutathione Peroxidase 1, 2, 3, 4 (Gpx 1, 2, 3, 4), Thioredoxin Reductase 1, 2, 3 (TR 1, 2, 3), Selenoprotein t (Selt), Selenoprotein w (Selw), Selenoprotein k (Selk), selenoprotein x1 (Sepx1), and significantly increased (P<0.05) the mRNA levels of the rest of selenoproteins. Correlation analysis showed that Deiodinase 3 (Dio 3), Selenoprotein m (Selm), 15-kDa Selenoprotein (Selp15), Selenoprotein h (Selh), Selenoprotein u (Selu), Selenoprotein i (Seli), Selenoprotein n (Seln), Selenoprotein p1 (Sepp1), Selenoprotein o (Selo), Selenoprotein s (Sels), Selenoprotein synthetase 2 (Sels2) and Selenoprotein p (Selp) had a negative correlation with Txn, while the rest of selenoproteins had a positive correlation with Txn. Combined in vivo and in vitro we can know that hamper Txn expression can inhibit Gpx 1, 2, 3, 4, TR 1, 2, 3, Dio 1, 2, Selt, Selw, Selk, Sepx1, meanwhile, over expression the rest of selenoproteins. In conclusion, the different selenoproteins possess and exhibit distinct responses to silence of Txn in chicken cardiomyocytes.


Subject(s)
Gene Expression , Myocytes, Cardiac/metabolism , Selenoproteins/genetics , Thioredoxins/metabolism , Animals , Chickens , Gene Knockdown Techniques , Models, Animal , Myocardium/metabolism , RNA, Messenger/metabolism , Selenium/deficiency , Thioredoxins/genetics
11.
Cell Signal ; 38: 192-200, 2017 10.
Article in English | MEDLINE | ID: mdl-28734787

ABSTRACT

Thioredoxin (Txn) system is the most crucial antioxidant defense mechanism in cell consisting of Txn, thioredoxin reductase (TR) and Nicotinamide Adenine Dinucleotide Phosphate (NADPH). Perturbations in Txn system may compromise cell survival through oxidative stress induction. Metabolic activity of insulin plays important roles in fulfilling the stable and persistent demands of heart through glucose metabolism. However, the roles of Txn and Txn system in insulin modulated cardiac energy metabolism have been less reported. Therefore, to investigate the role of Txn in myocardial metabolism, we developed a Se-deficient chicken model (0.033mg/kg) for in-vivo and Txn knock down cardiomyocytes culture model (siRNA) for in-vitro studies. Quantitative real time PCR and western blotting was performed. Se deficiency suppressed Txn and TR in cardiac tissues. Significant increases in ROS (P<0.05) levels signify the onset of oxidative stress and in both models. Se deficiency-induced Txn suppression model and Txn knock down cardiomyocytes models significantly decreased (P<0.05), the mRNA and protein levels of insulin-like growth factors (IGF1, IGF2), IGF-binding proteins (IGFBP2, IGFBP4), insulin receptor (IR), insulin receptor substrates (IRS1, IRS2), and glucose transporters (GLUT1, GLUT3, GLUT8), however, IGFBP3 expression increased in Txn knock down cardiomyocytes. In addition, in contrast to their respective controls, Se deficiency-induced Txn depleted tissues and Txn deleted cardiomyocytes showed suppression in mRNA and protein levels of PI3K, AKT, P-PI3K, and repression in FOX, P-FOX JNK genes. Combing the in vitro and in vivo experiments, we demonstrate that Txn gene suppression can cause dysfunction of insulin-modulated cardiac energy metabolism and increase insulin resistance through PI3K-Akt pathway inhibition. Herein, we conclude that inactivation of Txn system can alter cellular insulin response through IRS/PI3K/Akt pathway repression and JNK and FOX expression. These findings point out that Txn system can redox regulate the insulin dependent glucose metabolism in heart and is essential for cell vitality. Moreover, the increased expression of IGFBP3 indicates that it can be a potential negative modulator of metabolic activity of insulin in Txn deficient cells.


Subject(s)
Gene Knockdown Techniques , Insulin/pharmacology , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Selenium/deficiency , Thioredoxins/metabolism , Animals , Chickens , Gene Expression Regulation/drug effects , Myocardium/metabolism , Myocytes, Cardiac/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Thioredoxins/genetics
12.
J Inorg Biochem ; 170: 17-25, 2017 05.
Article in English | MEDLINE | ID: mdl-28214429

ABSTRACT

Dietary selenium (Se) deficiency can cause heart dysfunction, however the exact mechanism remains unclear. To understand this mechanism, 180day-old chicks, divided into two groups, C (control group) and L (low Se group), were fed with either a Se-sufficient (0.23mg/kg) or Se-deficient (0.033mg/kg) diets for 25days, respectively. Heart tissues and blood samples were collected. In L group, the activities of serum creatine kinase (CK) and creatine kinase-myoglobin (CK-MB) increased and typical ultrastructural apoptotic features were observed. Se deficiency up-regulated the mRNA levels of Cysteinyl aspartate specific proteinase 3 (Caspase-3), Cysteinyl aspartate specific proteinase 8 (Caspase-8), Cysteinyl aspartate specific proteinase 9 (Caspase-9), B cell lymphoma/leukemia 2 (Bcl-2), Bcl-2 Associated X Protein (Bax), (P<0.05), whereas, the mRNA levels of Microtubuleassociated protein light chains 3-1 (LC3-1), Autophagy associated gene 5 (ATG-5), Mammalian target of rapamycin (mTOR), Dynein and Becline-1 were down-regulated (P<0.05). Noticeably, Microtubuleassociated protein light chains 3-2 (LC3-2) mRNA level increased (P<0.05) by 20%. Western blot results showed that Se deficiency decreased the expression of Becline-1 and LC3-1 protein, however, the expression of Bax, Caspase-3 and Cysteinyl aspartate specific proteinase 12 (Caspase-12) increased at protein levels. The present study revealed that Se deficiency induced apoptosis while inhibited autophagy in chicken cardiomyocytes through Bax/Bcl-2 inhibition and caspases-mediated cleavage of Becline-1. Moreover, correlation analysis illustrates that apoptosis and autophagy might function contradictorily. Altogether we conclude that Se deficient chicken cardiomyocytes experienced apoptosis rather than autophagy which is considered to be more pro-survival.


Subject(s)
Apoptosis , Autophagy , Chickens/metabolism , Myocytes, Cardiac/metabolism , Selenium/deficiency , Animals , Apoptosis Regulatory Proteins/metabolism , Avian Proteins/metabolism , Muscle Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL