Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 45(Pt 3): 434-444, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28578007

ABSTRACT

Pyrroloquinoline quinone (PQQ) is a factor influencing on the mitochondrial biogenesis. In this study the PQQ effect on viability, total cell number, antioxidant capacity, mitochondrial biogenesis and differentiation potential was investigated in human induced Pluripotent Stem Cells (iPSC) - derived: neural stem cells (NSC), early neural progenitors (eNP) and neural progenitors (NP). Here we demonstrated that sensitivity to PQQ is dependent upon its dose and neural stage of development. Induction of the mitochondrial biogenesis by PQQ at three stages of neural differentiation was evaluated at mtDNA, mRNA and protein level. Changes in NRF1, TFAM and PPARGC1A gene expression were observed at all developmental stages, but only at eNP were correlated with the statistically significant increase in the mtDNA copy numbers and enhancement of SDHA, COX-1 protein level. Thus, the "developmental window" of eNP for PQQ-evoked mitochondrial biogenesis is proposed. This effect was independent of high antioxidant capacity of PQQ, which was confirmed in all tested cell populations, regardless of the stage of hiPSC neural differentiation. Furthermore, a strong induction of GFAP, with down regulation of MAP2 gene expression upon PQQ treatment was observed. This indicates a possibility of shifting the balance of cell differentiation in the favor of astroglia, but more research is needed at this point.


Subject(s)
Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects , PQQ Cofactor/pharmacology , Antioxidants/metabolism , Cell Count , Cell Differentiation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Dosage , Glial Fibrillary Acidic Protein/biosynthesis , Humans , Membrane Potential, Mitochondrial , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Mitochondria , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Reactive Oxygen Species/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
2.
Oncotarget ; 8(10): 16401-16413, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28146428

ABSTRACT

Male fertility disorders play a key role in half of all infertility cases. Reduction in testosterone induced by hypoxia might cause diseases in reproductive system and other organs. Hypoxic exposure caused a significant decrease of NRF1. Software analysis reported that the promoter region of steroidogenic acute regulatory protein (StAR) contained NRF1 binding sites, indicating NRF1 promoted testicular steroidogenesis. The purpose of this study is to determine NRF1 is involved in testosterone synthesis; and under hypoxia, the decrease of testosterone synthesis is caused by lower expression of NRF1. We designed both in vivo and in vitro experiments. Under hypoxia, the expressions of NRF1 in Leydig cells and testosterone level were significantly decreased both in vivo and in vitro. Overexpression and interference NRF1 could induced StAR and testosterone increased and decreased respectively. ChIP results confirmed the binding of NRF1 to StAR promoter region. In conclusion, decline of NRF1 expression downregulated the level of StAR, which ultimately resulted in a reduction in testosterone synthesis.


Subject(s)
Hypoxia/metabolism , Leydig Cells/metabolism , Nuclear Respiratory Factor 1/metabolism , Phosphoproteins/biosynthesis , Testosterone/biosynthesis , Animals , Cell Hypoxia/physiology , Down-Regulation , Humans , Hypoxia/genetics , Male , Mice , Mice, Inbred BALB C , Nuclear Respiratory Factor 1/biosynthesis , Oligonucleotide Array Sequence Analysis , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Promoter Regions, Genetic , Transfection
3.
Mol Med Rep ; 13(5): 4372-8, 2016 May.
Article in English | MEDLINE | ID: mdl-27035420

ABSTRACT

Nuclear respiratory factor 1 (NRF-1) has important roles in the regulation of several key metabolic genes required for cellular growth and respiration. A previous study by our group indicated that NRF­1 is involved in mitochondrial dysfunction induced by the environmental pollutant benzo[a]pyrene in the 16HBE human bronchial epithelial cell line. In the present study, it was observed that its genotoxic metabolite, anti­benzopyrene­7,8­diol­9,10­epoxide (BPDE), triggered cell cycle arrest in S­phase in 16HBE cells by activating ataxia-telangiectasia (ATM)/checkpoint kinase (Chk)2 and ATM and Rad3 related (ATR)/Chk1 signaling pathways. NRF­1 expression was suppressed by BPDE after treatment for 6 h. Flow cytometric analysis revealed that NRF­1 overexpression attenuated cell cycle arrest in S­phase induced by BPDE. In line with this result, DNA­damage checkpoints were activated following NRF­1 overexpression, as demonstrated by increased phosphorylation of ATM, Chk2 and γH2AX, but not ATR and Chk1, according to western blot analysis. It was therefore indicated that NRF­1 overexpression attenuated BPDE­induced S­phase arrest via the ATM/Chk2 signaling pathway.


Subject(s)
Bronchi/metabolism , Dihydroxydihydrobenzopyrenes/toxicity , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Nuclear Respiratory Factor 1/biosynthesis , Respiratory Mucosa/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Bronchi/pathology , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Nuclear Respiratory Factor 1/genetics , Respiratory Mucosa/pathology , S Phase Cell Cycle Checkpoints/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Int J Mol Med ; 37(2): 475-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26647788

ABSTRACT

Dojuksan is a traditional herbal medicine used in Korea and China to treat urinary diseases. In the present study, we aimed to examine the anti-inflammatory effects of an ethanol solvent extract of Dojuksan and a fraction (by bioassay-guided fractionation) derived from this extract, and to elucidate the specific mechanisms involved. The Dojuksan 30% ethanol extract (DEE) had a more significant and potent anti-inflammatory effect than the Dojuksan water extract (DWE). DEE markedly inhibited the production of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), as well as nuclear factor-κB (NF-κB) binding activity. We found that the anti-inflammatory effects of DEE were mediated by the induction of nuclear factor E2-related factor 2 (Nrf2)-dependent heme oxygenase-1 (HO-1). To further explore the anti-inflammatory effects of DEE, we generated 6 different fractions of DEE. Of these, DEE-5 decreased the production of NO more significantly than the other fractions. DEE-5 also significantly decreased the expression of iNOS and COX-2, and the production of NO, PGE2, TNF-α and IL-1ß. In addition, DEE-5 also significantly increased HO-1 levels; HO-1 significanlty contributed to the inhibitory effects of DEE-5 on the production of pro-inflammatory mediators. In this study, we determined whether the choice of extraction solvent affects the biological activity of Dojuksan, a traditional herbal formula. Our findings demonstrate that DEE and a fraction derived from this extract exerts anti-inflammatory effects through Nrf2­dependent HO-1 expression, and that DEE may thus have greater potential therapeutic application than DWE.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Heme Oxygenase-1/biosynthesis , Inflammation/drug therapy , Nuclear Respiratory Factor 1/biosynthesis , Animals , Cell Line , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Drugs, Chinese Herbal/chemistry , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Interleukin-1beta/biosynthesis , Lipopolysaccharides/toxicity , Mice , NF-kappa B/biosynthesis , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nuclear Respiratory Factor 1/genetics
5.
Sci Rep ; 5: 12983, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26268886

ABSTRACT

Upon translation, the N-terminal homology box 1 (NHB1) signal anchor sequence of Nrf1 integrates it within the endoplasmic reticulum (ER) whilst its transactivation domains [TADs, including acidic domain 1 (AD1), the flanking Asn/Ser/Thr-rich (NST) domain and AD2] are transiently translocated into the ER lumen, whereupon the NST domain is glycosylated to yield an inactive 120-kDa glycoprotein. Subsequently, these TADs are retrotranslocated into extra-luminal subcellular compartments, where Nrf1 is deglycosylated to yield an active 95-kDa isoform. Herein, we report that AD1 and AD2 are required for the stability of the 120-kDa Nrf1 glycoprotein, but not that of the non-glycosylated/de-glycosylated 95-kDa isoform. Degrons within AD1 do not promote proteolytic degradation of the 120-kDa Nrf1 glycoprotein. However, repositioning of AD2-adjoining degrons (i.e. DSGLS-containing SDS1 and PEST2 sequences) into the cyto/nucleoplasm enables selective topovectorial processing of Nrf1 by the proteasome and/or calpains to generate a cleaved active 85-kDa Nrf1 or a dominant-negative 36-kDa Nrf1γ. Production of Nrf1γ is abolished by removal of SDS1 or PEST2 degrons, whereas production of the cleaved 85-kDa Nrf1 is blocked by deletion of the ER luminal-anchoring NHB2 sequence (aa 81-106). Importantly, Nrf1 activity is positively and/or negatively regulated by distinct doses of proteasome and calpain inhibitors.


Subject(s)
Calpain/genetics , Nuclear Respiratory Factor 1/genetics , Protein Isoforms/genetics , Protein Processing, Post-Translational/genetics , Transcriptional Activation/genetics , Amino Acid Sequence/genetics , Animals , COS Cells , Calpain/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Gene Expression Regulation , Humans , Nuclear Respiratory Factor 1/biosynthesis , Proteasome Endopeptidase Complex/genetics , Protein Isoforms/biosynthesis , Proteolysis , Sequence Homology, Amino Acid
6.
Hum Mol Genet ; 24(16): 4516-29, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25976310

ABSTRACT

Ataxia oculomotor apraxia type 1 (AOA1) is an autosomal recessive disease caused by mutations in APTX, which encodes the DNA strand-break repair protein aprataxin (APTX). CoQ10 deficiency has been identified in fibroblasts and muscle of AOA1 patients carrying the common W279X mutation, and aprataxin has been localized to mitochondria in neuroblastoma cells, where it enhances preservation of mitochondrial function. In this study, we show that aprataxin deficiency impairs mitochondrial function, independent of its role in mitochondrial DNA repair. The bioenergetics defect in AOA1-mutant fibroblasts and APTX-depleted Hela cells is caused by decreased expression of SDHA and genes encoding CoQ biosynthetic enzymes, in association with reductions of APE1, NRF1 and NRF2. The biochemical and molecular abnormalities in APTX-depleted cells are recapitulated by knockdown of APE1 in Hela cells and are rescued by overexpression of NRF1/2. Importantly, pharmacological upregulation of NRF1 alone by 5-aminoimidazone-4-carboxamide ribonucleotide does not rescue the phenotype, which, in contrast, is reversed by the upregulation of NRF2 by rosiglitazone. Accordingly, we propose that the lack of aprataxin causes reduction of the pathway APE1/NRF1/NRF2 and their target genes. Our findings demonstrate a critical role of APTX in transcription regulation of mitochondrial function and the pathogenesis of AOA1 via a novel pathomechanistic pathway, which may be relevant to other neurodegenerative diseases.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-Binding Proteins/deficiency , Down-Regulation , Fibroblasts/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/biosynthesis , Nuclear Proteins/deficiency , Nuclear Respiratory Factor 1/biosynthesis , Signal Transduction , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Male , Mitochondria/pathology , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics
7.
Iran Biomed J ; 19(1): 23-8, 2015.
Article in English | MEDLINE | ID: mdl-25605486

ABSTRACT

BACKGROUND: The aim of the current study was to assess the mRNA levels of two mitochondria-related genes, including nuclear-encoded NRF1 (nuclear respiratory factor 1), mitochondrial transcription factor A (TFAM), and mitochondrial-encoded cytochrome c oxidase subunit 1 (MT-CO1) genes in various stages of the human oocyte maturation. METHODS: Oocytes were obtained from nine infertile women with male factor undergoing in vitro fertilization (IVF)/intra-cytoplasmic sperm injection protocol. Mitochondrial-related mRNA levels were performed by single-cell TaqMan real-time PCR. RESULTS: the expression level of the target genes was low at the germinal vesicle stage (P>0.05). Although the mRNA level of NRF1gene remained stable in metaphase I, the mRNA level of TFAM and MT-CO1 increased significantly (P<0.05).In metaphase II, the expression level of all genes increased compared to metaphase I (P<0.05). CONCLUSION: The overexpression levels of NRF1, TFAM, and MT-CO1 genes are related to the oocyte maturation. Therefore, the current study could be used clinically to improve the success rate of IVF.


Subject(s)
DNA-Binding Proteins/genetics , Electron Transport Complex IV/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Nuclear Respiratory Factor 1/genetics , Oocytes/cytology , Transcription Factors/genetics , Adult , DNA, Mitochondrial/genetics , DNA-Binding Proteins/biosynthesis , Electron Transport Complex IV/biosynthesis , Female , Fertilization in Vitro , Gene Expression Profiling , Humans , Infertility, Female , Male , Metaphase/genetics , Mitochondrial Proteins/biosynthesis , Nuclear Respiratory Factor 1/biosynthesis , Oocytes/metabolism , RNA, Messenger/genetics , RNA, Mitochondrial , Transcription Factors/biosynthesis , Transcription, Genetic/genetics , Young Adult
8.
Dev Cell ; 29(4): 392-405, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24871946

ABSTRACT

During development, major metabolic changes occur as cells become more specialized within a lineage. In the case of skeletal muscle, differentiation is accompanied by a switch from a glycolytic proliferative progenitor state to an oxidative postmitotic differentiated state. Such changes require extensive mitochondrial biogenesis leading to increased reactive oxygen species (ROS) production that needs to be balanced by an antioxidant system. Our analysis of double conditional Pitx2/3 mouse mutants, both in vivo during fetal myogenesis and ex vivo in primary muscle cell cultures, reveals excessive upregulation of ROS levels leading to DNA damage and apoptosis of differentiating cells. This is a consequence of downregulation of Nrf1 and genes for antioxidant enzymes, direct targets of Pitx2/3, leading to decreased expression of antioxidant enzymes, as well as impairment of mitochondrial function. Our analysis identifies Pitx2 and Pitx3 as key regulators of the intracellular redox state preventing DNA damage as cells undergo differentiation.


Subject(s)
Homeodomain Proteins/genetics , Nuclear Respiratory Factor 1/genetics , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cells, Cultured , DNA Damage/genetics , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Muscle Development/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Mutation , Nuclear Respiratory Factor 1/biosynthesis , Oxidation-Reduction , Up-Regulation , Homeobox Protein PITX2
9.
Int J Oncol ; 44(4): 1365-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24534923

ABSTRACT

Endocrine therapies have been successfully used for breast cancer patients with estrogen receptor α (ERα) positive tumors, but ~40% of patients relapse due to endocrine resistance. ß-glucans are components of plant cell walls that have immunomodulatory and anticancer activity. The objective of this study was to examine the activity of ß-D-glucan, purified from barley, in endocrine-sensitive MCF-7 versus endocrine-resistant LCC9 and LY2 breast cancer cells. ß-D-glucan dissolved in DMSO but not water inhibited MCF-7 cell proliferation in a concentration-dependent manner as measured by BrdU incorporation with an IC50 of ~164 ± 12 µg/ml. ß-D-glucan dissolved in DMSO inhibited tamoxifen/endocrine-resistant LCC9 and LY2 cell proliferation with IC50 values of 4.6 ± 0.3 and 24.2 ± 1.4 µg/ml, respectively. MCF-10A normal breast epithelial cells showed a higher IC50 ~464 µg/ml and the proliferation of MDA-MB-231 triple negative breast cancer cells was not inhibited by ß-D-glucan. Concentration-dependent increases in the BAX/BCL2 ratio and cell death with ß-D-glucan were observed in MCF-7 and LCC9 cells. PCR array analysis revealed changes in gene expression in response to 24-h treatment with 10 or 50 µg/ml ß-D-glucan that were different between MCF-7 and LCC9 cells as well as differences in basal gene expression between the two cell lines. Select results were confirmed by quantitative real-time PCR demonstrating that ß-D-glucan increased RASSF1 expression in MCF-7 cells and IGFBP3, CTNNB1 and ERß transcript expression in LCC9 cells. Our data indicate that ß-D-glucan regulates breast cancer-relevant gene expression and may be useful for inhibiting endocrine-resistant breast cancer cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Gene Expression/drug effects , Triple Negative Breast Neoplasms/drug therapy , beta-Glucans/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/biosynthesis , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , MCF-7 Cells , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , Tumor Suppressor Proteins/biosynthesis , beta Catenin/biosynthesis
10.
Free Radic Biol Med ; 65: 1435-1446, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24120971

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of the insulin signaling pathway and is considered a promising therapeutic target in the treatment of diabetes. However, the role of PTP1B in palmitate-induced mitochondrial dysfunction and apoptosis in skeletal muscle cells has not been studied. Here we investigate the effects of PTP1B modulation on mitochondrial function and apoptosis and elucidate the underlying mechanisms in skeletal muscle cells. PTP1B inhibition significantly reduced palmitate-induced mitochondrial dysfunction and apoptosis in C2C12 cells, as these cells had increased expression levels of PGC-1α, Tfam, and NRF-1; enhanced ATP level and cellular viability; decreased TUNEL-positive cells; and decreased caspase-3 and -9 activity. Alternatively, overexpression of PTP1B resulted in mitochondrial dysfunction and apoptosis in these cells. PTP1B silencing improved mitochondrial dysfunction by an increase in the expression of SIRT1 and a reduction in the phosphorylation of p65 NF-κB. The protection from palmitate-induced apoptosis by PTP1B inhibition was also accompanied by a decrease in protein level of serine palmitoyl transferase, thus resulting in lower ceramide content in muscle cells. Exogenous addition of C2-ceramide to PTP1B-knockdown cells led to a reduced generation of reactive oxygen species (ROS), whereas PTP1B overexpression demonstrated an elevated ROS production in myotubes. In addition, PTP1B inhibition was accompanied by decreased JNK phosphorylation and increased insulin-stimulated Akt (Ser473) phosphorylation, whereas overexpression of PTP1B had the opposite effect. The overexpression of PTP1B also induced the nuclear localization of FOXO-1, but in contrast, suppression of PTP1B reduced palmitate-induced nuclear localization of FOXO-1. In summary, our results indicate that PTP1B modulation results in (1) alterations in mitochondrial function by changes in the activity of SIRT1/NF-κB/PGC-1α pathways and (2) changes in apoptosis that result from either a direct effect of PTP1B on the insulin signaling pathway or an indirect influence on ceramide content, ROS generation, JNK activation, and FOXO-1 nuclear translocation.


Subject(s)
Apoptosis/physiology , Insulin/metabolism , Mitochondria/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Active Transport, Cell Nucleus , Adenosine Triphosphate/biosynthesis , Apoptosis/genetics , Caspase 3/biosynthesis , Caspase 9/biosynthesis , Cell Line , Cell Survival , DNA-Binding Proteins/biosynthesis , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondrial Proteins/biosynthesis , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Nuclear Respiratory Factor 1/biosynthesis , Palmitates/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species , Signal Transduction/drug effects , Sirtuin 1/biosynthesis , Sirtuin 1/genetics , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Transcription Factor RelA/metabolism , Transcription Factors/biosynthesis
11.
Pharmazie ; 68(4): 270-3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23700793

ABSTRACT

This study aimed to investigate the effect of pioglitazone (PIO) on insulin secretion and mitochondrial ultrastructure and function in 3 cells. HIT-T15 cells were treated with control or palmitate (free fat acids, FFA) or/and PIO and divided into 7 groups: Control group; 0.5 mmol/l FFA (LF); 0.5 mmol/l FFA plus 10-7 mol/I PIO (LFLP); 0.5 mmol/l FFA plus 10-5mol/I PIO (LFHP); 1.0 mmol/l FFA (HF); 1.0 mmol/l FFA plus 10-7mol/I PIO (HFLP); 1.0 mmol/l FFA plus 10-5 mol/I PIO (HFHP). Apoptotic peaks, mitochondrial ultrastructure, ATP/ADP, mRNA levels of peroxisome proliferater activated receptor gamma coactivator-1 (PGC-1) and nucleus respiratory factor-1 (NRF-1) as well as insulin secretion were measured. The results showed that palmitate impaired mitochondrion structure, which could be alleviated by PIO. Palmitate could increase apoptotic peaks, decrease ATP/ADP ratio, enhance the expression of PGC-1 mRNA and NRF-1 mRNA, and decrease glucose stimulated insulin secretion (GSIS). In contrast, PIO could decrease apoptotic peaks, restore partly ATP/ADP ratio, decrease the expression of PGC-1 mRNA and NRF-1 mRNA, and increase GSIS level. These results demonstrate that PIO could ameliorate palmitate induced damage to mitochondrion ultrastructure and function and restore GSIS, accompanied by the modulation of PGC-1 and NRF-1 expression. These findings provide new insight into the hypoglycemic effects of PIO and help develop new agents for diabetes therapy.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mitochondria/ultrastructure , Palmitates/antagonists & inhibitors , Palmitates/toxicity , Thiazolidinediones/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Line , Chromatography, High Pressure Liquid , Flow Cytometry , Heat-Shock Proteins/biosynthesis , Humans , In Situ Nick-End Labeling , Indicators and Reagents , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/ultrastructure , Microscopy, Electron , Mitochondria/drug effects , Nuclear Respiratory Factor 1/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pioglitazone , Real-Time Polymerase Chain Reaction , Transcription Factors/biosynthesis
12.
Diabetes ; 60(8): 2051-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21677280

ABSTRACT

OBJECTIVE: Aging increases the risk of developing impaired glucose tolerance (IGT) and type 2 diabetes. It has been proposed that increased reactive oxygen species (ROS) generation by dysfunctional mitochondria could play a role in the pathogenesis of these metabolic abnormalities. We examined whether aging per se (in subjects with normal glucose tolerance [NGT]) impairs mitochondrial function and how this relates to ROS generation, whether older subjects with IGT have a further worsening of mitochondrial function (lower ATP production and elevated ROS generation), and whether exercise reverses age-related changes in mitochondrial function. RESEARCH DESIGN AND METHODS: Mitochondrial ATP and ROS production were measured in muscle from younger individuals with NGT, older individuals with NGT, and older individuals with IGT. Measurements were performed before and after 16 weeks of aerobic exercise. RESULTS: ATP synthesis was lower in older subjects with NGT and older subjects with IGT versus younger subjects. Notably, mitochondria from older subjects (with NGT and IGT) displayed reduced ROS production versus the younger group. ATP and ROS production were similar between older groups. Exercise increased ATP synthesis in the three groups. Mitochondrial ROS production also increased after training. Proteomic analysis revealed downregulation of several electron transport chain proteins with aging, and this was reversed by exercise. CONCLUSIONS: Old mitochondria from subjects with NGT and IGT display mitochondrial dysfunction as manifested by reduced ATP production but not with respect to increased ROS production. When adjusted to age, the development of IGT in elderly individuals does not involve changes in mitochondrial ATP and ROS production. Lastly, exercise reverses the mitochondrial phenotype (proteome and function) of old mitochondria.


Subject(s)
Adenosine Triphosphate/biosynthesis , Aging/physiology , Glucose Intolerance/physiopathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Adolescent , Adult , Aged , DNA-Binding Proteins , Exercise , Gene Expression Profiling , Heat-Shock Proteins/biosynthesis , Humans , Lipid Peroxidation , Mitochondrial Proteins , Nuclear Respiratory Factor 1/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proteomics , Transcription Factors/biosynthesis
13.
J Biol Chem ; 286(12): 10605-17, 2011 03 25.
Article in English | MEDLINE | ID: mdl-21245132

ABSTRACT

Endurance exercise is known to induce metabolic adaptations in skeletal muscle via activation of the transcriptional co-activator peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α). PGC-1α regulates mitochondrial biogenesis via regulating transcription of nuclear-encoded mitochondrial genes. Recently, PGC-1α has been shown to reside in mitochondria; however, the physiological consequences of mitochondrial PGC-1α remain unknown. We sought to delineate if an acute bout of endurance exercise can mediate an increase in mitochondrial PGC-1α content where it may co-activate mitochondrial transcription factor A to promote mtDNA transcription. C57Bl/6J mice (n = 12/group; ♀ = ♂) were randomly assigned to sedentary (SED), forced-endurance (END) exercise (15 m/min for 90 min), or forced endurance +3 h of recovery (END+3h) group. The END group was sacrificed immediately after exercise, whereas the SED and END+3h groups were euthanized 3 h after acute exercise. Acute exercise coordinately increased the mRNA expression of nuclear and mitochondrial DNA-encoded mitochondrial transcripts. Nuclear and mitochondrial abundance of PGC-1α in END and END+3h groups was significantly higher versus SED mice. In mitochondria, PGC-1α is in a complex with mitochondrial transcription factor A at mtDNA D-loop, and this interaction was positively modulated by exercise, similar to the increased binding of PGC-1α at the NRF-1 promoter. We conclude that in response to acute altered energy demands, PGC-1α re-localizes into nuclear and mitochondrial compartments where it functions as a transcriptional co-activator for both nuclear and mitochondrial DNA transcription factors. These results suggest that PGC-1α may dynamically facilitate nuclear-mitochondrial DNA cross-talk to promote net mitochondrial biogenesis.


Subject(s)
Cell Nucleus/metabolism , Energy Metabolism/physiology , Mitochondria, Muscle/metabolism , Physical Conditioning, Animal , Trans-Activators/biosynthesis , Transcription, Genetic/physiology , Animals , Cell Nucleus/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Male , Mice , Mitochondria, Muscle/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Endurance/physiology , Promoter Regions, Genetic/physiology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Exp Neurol ; 220(2): 328-34, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19766117

ABSTRACT

Selenoprotein H (SelH) is one of the 25 so far identified selenoproteins. Selenoproteins may function as antioxidants, heavy metal antidotes, and neural survival factors. Previous studies have shown that overexpression of SelH in HT22 cells protected the cells from UVB irradiation-induced death by reducing superoxide formation. The objective of this study was to determine the effects of SelH on cell signaling pathways after UVB irradiation. We exposed both human SelH- and vector-transfected HT22 cells to UVB irradiation and collected samples at 5 and 17 h of recovery. Cell viability was assessed, as well as protein levels of caspase-3, -8, -9, apoptosis-inducing factor (AIF), P53, nuclear respiratory factor-1 (NRF-1) and heat shock protein 40 (HSP40). Mitochondrial membrane potential was determined by flow cytometry. Overexpression of SelH protected cells against UVB-induced injury by blockade of the mitochondria-initiated cell death pathway, prevention of mitochondrial membrane depolarization, and suppression of the increase of p53. Furthermore, overexpression of SelH increased levels of NRF-1, an antioxidant, and HSP40, a protein chaperone that repairs denatured protein. We conclude that SelH protects neurons against UVB-induced damage by inhibiting apoptotic cell death pathways, by preventing mitochondrial depolarization, and by promoting cell survival pathways.


Subject(s)
DNA-Binding Proteins/physiology , Neurons/metabolism , Neurons/radiation effects , Selenoproteins/physiology , Signal Transduction/genetics , Signal Transduction/radiation effects , Ultraviolet Rays , Animals , Apoptosis/genetics , Apoptosis/physiology , Apoptosis/radiation effects , Blotting, Western , Caspase 3/metabolism , Caspase 3/radiation effects , Caspase 9/metabolism , Caspase 9/radiation effects , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Cell Survival/radiation effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression/physiology , Genetic Vectors , HSP40 Heat-Shock Proteins/biosynthesis , HSP40 Heat-Shock Proteins/genetics , Humans , Membrane Potentials/physiology , Mice , Mitochondrial Membranes/physiology , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , Selenoproteins/biosynthesis , Selenoproteins/genetics , Tumor Suppressor Protein p53
15.
Toxicology ; 264(1-2): 10-5, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19616056

ABSTRACT

Perfluorooctanoic acid (PFOA), used in the production of non-stick surface compounds, exhibits a worldwide distribution in the serum of humans and wildlife. In rodents PFOA transactivates PPARalpha and PPARgamma nuclear receptors and increases mitochondrial DNA (mtDNA) copy number, which may be critical to the altered metabolic state of affected animals. A key regulator of mitochondrial biogenesis and transcription of mitochondrial genes is the PPARgamma coactivator-1alpha (Pgc-1alpha) protein. The purpose of this study was to determine if Pgc-1alpha is implicated in the stimulation of mitochondrial biogenesis that occurs following the treatment of rats with PFOA. Livers from adult male Sprague-Dawley rats that received a 30 mg/kg daily oral dose of PFOA for 28 days were used for all experiments. Analysis of mitochondrial replication and transcription was performed by real time PCR, and proteins were detected using western blotting. PFOA treatment caused a transcriptional activation of the mitochondrial biogenesis pathway leading to a doubling of mtDNA copy number. Further, transcription of OXPHOS genes encoded by mtDNA was 3-4 times greater than that of nuclear encoded genes, suggestive of a preferential induction of mtDNA transcription. Western blot analysis revealed an increase in Pgc-1alpha, unchanged Tfam and decreased Cox II and Cox IV subunit protein expression. We conclude that PFOA treatment in rats induces mitochondrial biogenesis at the transcriptional level with a preferential stimulation of mtDNA transcription and that this occurs by way of activation of the Pgc-1alpha pathway. Implication of the Pgc-1alpha pathway is consistent with PPARgamma transactivation by PFOA and reveals new understanding and possibly new critical targets for assessing or averting the associated metabolic disease.


Subject(s)
Caprylates/pharmacology , Fluorocarbons/pharmacology , Mitochondria, Liver/drug effects , Transcription, Genetic/drug effects , Animals , Blotting, Western , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA, Mitochondrial/biosynthesis , DNA, Mitochondrial/isolation & purification , Electron Transport/drug effects , Male , Mitochondria, Liver/metabolism , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , PPAR gamma/biosynthesis , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , RNA/biosynthesis , RNA/isolation & purification , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription Factors/biosynthesis , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/physiology
16.
Mol Endocrinol ; 22(3): 609-22, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18048642

ABSTRACT

Estrogen has direct and indirect effects on mitochondrial activity, but the mechanisms mediating these effects remain unclear. Others reported that long-term estradiol (E(2)) treatment increased nuclear respiratory factor-1 (NRF-1) protein in cerebral blood vessels of ovariectomized rats. NRF-1 is a transcription factor that regulates the expression of nuclear-encoded mitochondrial genes, e.g. mitochondrial transcription factor A (TFAM), that control transcription of the mitochondrial genome. Here we tested the hypothesis that E(2) increases NRF-1 transcription resulting in a coordinate increase in the expression of nuclear- and mitochondrial- encoded genes and mitochondrial respiratory activity. We show that E(2) increased NRF-1 mRNA and protein in MCF-7 breast and H1793 lung adenocarcinoma cells in a time-dependent manner. E(2)-induced NRF-1 expression was inhibited by the estrogen receptor (ER) antagonist ICI 182,780 and actinomycin D but not by phosphoinositide-3 kinase and MAPK inhibitors, indicating a genomic mechanism of E(2) regulation of NRF-1 transcription. An estrogen response element (ERE) in the NRF-1 promoter bound ER alpha and ER beta in vitro, and E(2) induced ER alpha and ER beta recruitment to this ERE in chromatin immunoprecipitation assays in MCF-7 cells. The NRF-1 ERE activated reporter gene expression in transfected cells. Small interfering RNA to ER alpha and ER beta revealed that ER alpha mediates E(2)-induced NRF-1 transcription. The E(2)-induced increase in NRF-1 was followed by increased TFAM and the transcription of Tfam-regulated mitochondrial DNA-encoded COI and NDI genes and increased mitochondrial biogenesis. Knockdown of NRF-1 blocked E(2) stimulation of mitochondrial biogenesis and activity, indicating a mechanism by which estrogens regulate mitochondrial function by increasing NRF-1 expression.


Subject(s)
Estradiol/pharmacology , Mitochondria/drug effects , Nuclear Respiratory Factor 1/biosynthesis , Transcription, Genetic/physiology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA, Mitochondrial/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Fulvestrant , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Nitriles/pharmacology , Nuclear Respiratory Factor 1/genetics , Phenols , Promoter Regions, Genetic/drug effects , Propionates/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics
17.
Acta Pharmacol Sin ; 28(10): 1541-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17883938

ABSTRACT

AIM: To investigate the effect of icariin on the expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1alpha), peroxisome proliferator-activated receptor alpha (PPARalpha), and nuclear respiratory factor 1 (NRF-1) on cardiomyocyte differentiation of murine embryonic stem (ES) cells in vitro. METHODS: The cardiomyocytes derived from murine ES cells were verified by immunocytochemistry using confocal laser scanning microscopy. Cardiac-specific sarcomeric proteins (ie alpha-actinin, troponin T) were evaluated when embryoid bodies (EB) were treated with icariin or retinoid acid. The expression of PGC-1alpha, PPARalpha, and NRF-1 were analyzed using both semiquantitative RT-PCR and Western blotting in cardiomyocyte differentiation. The phosphorylation of the p38 mitogen-activated protein kinase (MAPK) was studied in the differentiation process, and its specific inhibitor SB203580 was employed to confirm the function of the p38 MAPK on icariin-induced cardiac differentiation. RESULTS: The application of icariin significantly induced the cardiomyocyte differentiation of EB as indicated by the promoted expression of alpha-actinin and troponin T. The expression of PGC-1alpha, PPARalpha, and NRF-1 increased coincidently in early differentiation and the increase was dose-dependently upregulated by icariin treatment. The phosphorylation of the p38 MAPK peaked on d 6 and decreased after d 8, and the activation was further enhanced and prolonged when the EB were subjected to icariin, which was concurrent with the elevation of PGC-1alpha, PPARalpha, and NRF-1. Moreover, the inhibition of the p38 MAPK pathway by SB203580 efficiently abolished icariin-stimulated cardiomyocyte differentiation and resulted in the capture of the upregulation of PGC-1alpha, PPARalpha, and NRF-1. CONCLUSION: Taken together, icariin promoted the expression of PGC-1alpha, PPARalpha, and NRF-1 during cardiomyocyte differentiation of murine ES cells in vitro and the effect was partly responsible for the activation of the p38 MAPK.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Flavonoids/pharmacology , Myocytes, Cardiac/cytology , Transcription Factors/biosynthesis , Animals , Blotting, Western , Dose-Response Relationship, Drug , Embryonic Stem Cells/metabolism , Enzyme Inhibitors/pharmacology , Epimedium/chemistry , Flavonoids/administration & dosage , Flavonoids/isolation & purification , Imidazoles/pharmacology , Mice , Myocytes, Cardiac/metabolism , Nuclear Respiratory Factor 1/biosynthesis , Nuclear Respiratory Factor 1/genetics , PPAR alpha/biosynthesis , PPAR alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation/drug effects , Plants, Medicinal/chemistry , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Diabetes Metab Res Rev ; 22(6): 492-8, 2006.
Article in English | MEDLINE | ID: mdl-16752430

ABSTRACT

BACKGROUND: Physical exercise provides health benefits for people with type 2 diabetes mellitus, partly by enhancing skeletal muscle insulin action. We tested the hypothesis that changes in expression of key genes in skeletal muscles relate to exercise-induced improvements in type 2 diabetic patients. METHODS: We determined mRNA expression of 20 selected genes following a self-supervised program of walking (> 150 min per week) over a 4-month period. RESULTS: This level of physical activity improved clinical parameters in approximately half the participants, as determined by reduced hypertension and enhanced insulin sensitivity (defined by reduced plasma-insulin levels and improved homeostasis model assessment (HOMA)). Skeletal muscle mRNA expression of Cbl-associated protein (CAP), diacylglycerol kinase (DGK)delta, uncoupling protein (UCP) 3, nuclear respiratory factor (NRF)-1, and peroxisome proliferator-activated receptor (PPAR)delta tended to increase in type 2 diabetic patients with an improved clinical profile. Skeletal muscle protein expression of PPARdelta and UCP3 was increased significantly after physical exercise in patients with an improved clinical profile, but were unchanged in patients who did not show exercise-mediated improvements in clinical parameters. CONCLUSIONS: This study provides clinical evidence that improvements in insulin sensitivity can be achieved in type 2 diabetic patients after individually executed low-intensity exercise training. Moreover, the positive clinical response to exercise is correlated with changes in skeletal muscle proteins involved in the regulation of mitochondrial biogenesis and metabolism. These changes in skeletal muscle gene expression offer a possible molecular explanation for the improvements in clinical outcomes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Exercise/physiology , Ion Channels/biosynthesis , Mitochondrial Proteins/biosynthesis , Muscle, Skeletal/metabolism , PPAR delta/biosynthesis , Cytoskeletal Proteins/biosynthesis , Diabetes Mellitus, Type 2/genetics , Diacylglycerol Kinase/biosynthesis , Female , Gene Expression , Humans , Male , Middle Aged , Nuclear Respiratory Factor 1/biosynthesis , RNA, Messenger/metabolism , Uncoupling Protein 3
19.
Reprod Biol Endocrinol ; 3: 65, 2005 Nov 14.
Article in English | MEDLINE | ID: mdl-16285882

ABSTRACT

BACKGROUND: Recent work has shown that mitochondrial biogenesis and mitochondrial functions are critical determinants of embryonic development. However, the expression of the factors controlling mitochondrial biogenesis in early embryogenesis has received little attention so far. METHODS: We used real-time quantitative PCR to quantify mitochondrial DNA (mtDNA) in bovine oocytes and in various stages of in vitro produced embryos. To investigate the molecular mechanisms responsible for the replication and the transcriptional activation of mtDNA, we quantified the mRNA corresponding to the mtDNA-encoded cytochrome oxidase 1 (COX1), and two nuclear-encoded factors, i.e. the Nuclear Respiratory Factor 1 (NRF1), and the nuclear-encoded Mitochondrial Transcription Factor A (mtTFA). RESULTS: Unlike findings reported in mouse embryos, the mtDNA content was not constant during early bovine embryogenesis. We found a sharp, 60% decrease in mtDNA content between the 2-cell and the 4/8-cell stages. COX1 mRNA was constant until the morula stage after which it increased dramatically. mtTFA mRNA was undetectable in oocytes and remained so until the 8/16-cell stage; it began to appear only at the morula stage, suggesting de novo synthesis. In contrast, NRF1 mRNA was detectable in oocytes and the quantity remained constant until the morula stage. CONCLUSION: Our results revealed a reduction of mtDNA content in early bovine embryos suggesting an active process of mitochondrial DNA degradation. In addition, de novo mtTFA expression associated with mitochondrial biogenesis activation and high levels of NRF1 mRNA from the oocyte stage onwards argue for the essential function of these factors during the first steps of bovine embryogenesis.


Subject(s)
DNA, Mitochondrial/metabolism , DNA-Binding Proteins/biosynthesis , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Mitochondrial Proteins/biosynthesis , Nuclear Respiratory Factor 1/biosynthesis , Oocytes/metabolism , Transcription Factors/biosynthesis , Animals , Cattle , Electron Transport Complex IV/biosynthesis , Female , RNA, Messenger/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...