Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38979162

ABSTRACT

The liver, the largest internal organ and a metabolic hub, undergoes significant declines due to aging, affecting mitochondrial function and increasing the risk of systemic liver diseases. How the mitochondrial three-dimensional (3D) structure changes in the liver across aging, and the biological mechanisms regulating such changes confers remain unclear. In this study, we employed Serial Block Face-Scanning Electron Microscopy (SBF-SEM) to achieve high-resolution 3D reconstructions of murine liver mitochondria to observe diverse phenotypes and structural alterations that occur with age, marked by a reduction in size and complexity. We also show concomitant metabolomic and lipidomic changes in aged samples. Aged human samples reflected altered disease risk. To find potential regulators of this change, we examined the Mitochondrial Contact Site and Cristae Organizing System (MICOS) complex, which plays a crucial role in maintaining mitochondrial architecture. We observe that the MICOS complex is lost during aging, but not Sam50. Sam50 is a component of the sorting and assembly machinery (SAM) complex that acts in tandem with the MICOS complex to modulate cristae morphology. In murine models subjected to a high-fat diet, there is a marked depletion of the mitochondrial protein SAM50. This reduction in Sam50 expression may heighten the susceptibility to liver disease, as our human biobank studies corroborate that Sam50 plays a genetically regulated role in the predisposition to multiple liver diseases. We further show that changes in mitochondrial calcium dysregulation and oxidative stress accompany the disruption of the MICOS complex. Together, we establish that a decrease in mitochondrial complexity and dysregulated metabolism occur with murine liver aging. While these changes are partially be regulated by age-related loss of the MICOS complex, the confluence of a murine high-fat diet can also cause loss of Sam50, which contributes to liver diseases. In summary, our study reveals potential regulators that affect age-related changes in mitochondrial structure and metabolism, which can be targeted in future therapeutic techniques.

2.
Adv Biol (Weinh) ; 7(10): e2200202, 2023 10.
Article in English | MEDLINE | ID: mdl-37140138

ABSTRACT

Mitochondria respond to metabolic demands of the cell and to incremental damage, in part, through dynamic structural changes that include fission (fragmentation), fusion (merging of distinct mitochondria), autophagic degradation (mitophagy), and biogenic interactions with the endoplasmic reticulum (ER). High resolution study of mitochondrial structural and functional relationships requires rapid preservation of specimens to reduce technical artifacts coupled with quantitative assessment of mitochondrial architecture. A practical approach for assessing mitochondrial fine structure using two dimensional and three dimensional high-resolution electron microscopy is presented, and a systematic approach to measure mitochondrial architecture, including volume, length, hyperbranching, cristae morphology, and the number and extent of interaction with the ER is described. These methods are used to assess mitochondrial architecture in cells and tissue with high energy demand, including skeletal muscle cells, mouse brain tissue, and Drosophila muscles. The accuracy of assessment is validated in cells and tissue with deletion of genes involved in mitochondrial dynamics.


Subject(s)
Mitochondria , Mitochondrial Membranes , Mice , Animals , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Microscopy, Electron, Scanning , Cells, Cultured
3.
Cell Rep ; 38(8): 110408, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196497

ABSTRACT

The adipose tissue is a key site regulating energy metabolism. One of the contributing factors behind this is browning of white adipose tissue (WAT). However, knowledge of the intracellular determinants of the browning process remains incomplete. By generating adipocyte-specific Senp2 knockout (Senp2-aKO) mice, here we show that SENP2 negatively regulates browning by de-conjugating small ubiquitin-like modifiers from C/EBPß. Senp2-aKO mice are resistant to diet-induced obesity due to increased energy expenditure and heat production. Senp2 knockout promotes beige adipocyte accumulation in inguinal WAT by upregulation of thermogenic gene expression. In addition, SENP2 knockdown promotes thermogenic adipocyte differentiation of precursor cells isolated from inguinal and epididymal WATs. Mechanistically, sumoylated C/EBPß, a target of SENP2, suppresses expression of HOXC10, a browning inhibitor, by recruiting a transcriptional repressor DAXX. These findings indicate that a SENP2-C/EBPß-HOXC10 axis operates for the control of beige adipogenesis in inguinal WAT.


Subject(s)
Adipocytes, Beige , CCAAT-Enhancer-Binding Protein-beta , Cysteine Endopeptidases , Small Ubiquitin-Related Modifier Proteins , Adipocytes, Beige/metabolism , Adipogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cysteine Endopeptidases/metabolism , Energy Metabolism/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Thermogenesis/genetics
4.
Metabolism ; 95: 27-35, 2019 06.
Article in English | MEDLINE | ID: mdl-30902749

ABSTRACT

BACKGROUND AND PURPOSE: In addition to the central nervous system-mediated action, leptin also directly induces fatty acid oxidation in skeletal muscle. Rapid induction of FAO by leptin is mediated by the AMP-activated protein kinase (AMPK) pathway, but the mechanism of prolonged FAO by leptin was previously unknown. In an earlier study, we showed that free fatty acids increase transcription of small ubiquitin-like modifier (SUMO) specific protease 2 (SENP2) in skeletal muscle, and that SENP2 stimulates expression of FAO-associated enzymes by deSUMOylating peroxisome proliferator-activated receptors, PPARδ and PPARγ. In this study, we examine whether SENP2 is involved in prolonged stimulation of FAO by leptin. METHODS: The Effect of leptin on expression of SENP2 and on SENP2-mediated FAO was investigated by using western blotting and real time qPCR of C2C12 myotubes, and of C2C12 myotubes in which expression of specific genes was knocked down using siRNAs. Additionally, muscle-specific SENP2 knockout mice were generated to test the involvement of SENP2 in leptin-induced FAO in vivo. RESULTS: We show that leptin treatment of C2C12 myotubes causes signal transducer and activator of transcription 3 (STAT3) to bind to the Senp2 promoter, inducing SENP2 expression. We also show that leptin increases the binding of PPARδ and PPARγ to PPRE sites in the promoters of two FAO-associated genes: long-chain acyl-CoA synthetase 1 (Acsl1) or carnitine palmitoyl transferase 1b (Cpt1b). When SENP2 is knocked down in myotubes, leptin-induced expression of FAO-associated enzymes and prolonged increase of FAO are suppressed, but rapid increase of FAO is unaffected. In addition, leptin-induced expression of FAO-associated enzymes was not observed in muscle tissue of SENP2 knockout mice. CONCLUSIONS: We demonstrate that the peripheral actions of leptin on FAO are mediated by two different pathways: AMPK causes a rapid increase in FAO, and SENP2 of the STAT3 pathway causes a slow, prolonged increase in FAO.


Subject(s)
Cysteine Endopeptidases/metabolism , Fatty Acids/metabolism , Leptin/pharmacology , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Gene Knockdown Techniques , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Oxidation-Reduction
5.
PLoS One ; 12(10): e0186021, 2017.
Article in English | MEDLINE | ID: mdl-29016649

ABSTRACT

The small heterodimer partner (SHP) regulates fatty acid oxidation and lipogenesis in the liver by regulating peroxisome proliferator-activated receptor (PPAR) γ expression. SHP is also abundantly expressed in the myocardium. We investigated the effect of SHP expression on myocardia assessing not only heart structure and function but also lipid metabolism and related gene expression in a SHP deletion animal model. Transcriptional profiling with a microarray revealed that genes participating in cell growth, cytokine signalling, phospholipid metabolism, and extracellular matrix are up-regulated in the myocardia of SHP knockout (KO) mice compared to those of wild-type (WT) mice (nominal p value < 0.05). Consistent with these gene expression changes, the left ventricular masses of SHP KO mice were significantly higher than WT mice (76.8 ± 20.5 mg vs. 52.8 ± 6.8 mg, P = 0.0093). After 12 weeks of high fat diet (HFD), SHP KO mice gained less weight and exhibited less elevation in serum-free fatty acid and less ectopic lipid accumulation in the myocardium than WT mice. According to microarray analysis, genes regulated by PPARγ1 and PPARα were down-regulated in myocardia of SHP KO mice compared to their expression in WT mice after HFD, suggesting that the reduction in lipid accumulation in the myocardium resulted from a decrease in lipogenesis regulated by PPARγ. We confirmed the reduced expression of PPARγ1 and PPARα target genes such as CD36, medium-chain acyl-CoA dehydrogenase, long-chain acyl-CoA dehydrogenase, and very long-chain acyl-CoA dehydrogenase by SHP KO after HFD.


Subject(s)
Lipogenesis/genetics , Myocardium/metabolism , Obesity/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcriptome , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Congenital Bone Marrow Failure Syndromes , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Gene Expression Profiling , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , Myocardium/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Protective Factors , Receptors, Cytoplasmic and Nuclear/deficiency , Signal Transduction
6.
Diabetes Metab J ; 40(5): 406-413, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27535645

ABSTRACT

BACKGROUND: Panax ginseng has glucose-lowering effects, some of which are associated with the improvement in insulin resistance in skeletal muscle. Because mitochondria play a pivotal role in the insulin resistance of skeletal muscle, we investigated the effects of the ginsenoside Rg3, one of the active components of P. ginseng, on mitochondrial function and biogenesis in C2C12 myotubes. METHODS: C2C12 myotubes were treated with Rg3 for 24 hours. Insulin signaling pathway proteins were examined by Western blot. Cellular adenosine triphosphate (ATP) levels and the oxygen consumption rate were measured. The protein or mRNA levels of mitochondrial complexes were evaluated by Western blot and quantitative reverse transcription polymerase chain reaction analysis. RESULTS: Rg3 treatment to C2C12 cells activated the insulin signaling pathway proteins, insulin receptor substrate-1 and Akt. Rg3 increased ATP production and the oxygen consumption rate, suggesting improved mitochondrial function. Rg3 increased the expression of peroxisome proliferator-activated receptor γ coactivator 1α, nuclear respiratory factor 1, and mitochondrial transcription factor, which are transcription factors related to mitochondrial biogenesis. Subsequent increased expression of mitochondrial complex IV and V was also observed. CONCLUSION: Our results suggest that Rg3 improves mitochondrial function and the expression of key genes involved in mitochondrial biogenesis, leading to an improvement in insulin resistance in skeletal muscle. Rg3 may have the potential to be developed as an anti-hyperglycemic agent.

7.
Mol Cell Endocrinol ; 436: 50-8, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27452800

ABSTRACT

Thyroid-stimulating hormone (TSH) receptor is expressed in extrathyroidal tissues such as hepatocytes, adipocytes, and skeletal muscle, which suggests a possible novel role of TSH in various metabolic processes in extrathyroidal tissues independent of thyroid hormones. We investigated whether TSH has any effects on glucose tolerance and insulin sensitivity in the skeletal muscle using diet-induced obesity (DIO) mouse models and rodent skeletal muscle cells. TSH improved glucose tolerance in DIO mice and this was associated with an improvement of skeletal muscle insulin sensitivity resulting from the increased expression of insulin receptor substrate (IRS)-1 protein and mRNA therein. TSH significantly increased both basal and insulin-stimulated glucose transport in rat L6 myotubes and increased the expression of IRS-1 protein and mRNA in these cells as well. TSH also stimulated Irs1 promoter activation; this stimulation was abolished by protein kinase A (PKA) inhibition using H89 or by mutation of the cAMP-response element site located at -1155 to -875 bp of the Irs1 promoter region, supporting a novel role of TSH activated-cAMP/PKA/CREB signaling in the regulation of Irs1 expression. In conclusion, TSH improves insulin sensitivity in skeletal muscle by increasing Irs1 gene expression. This regulatory effect is mediated by a PKA-CREB-dependent pathway.


Subject(s)
Insulin Receptor Substrate Proteins/genetics , Insulin Resistance , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , Thyrotropin/pharmacology , Up-Regulation/drug effects , Animals , Base Sequence , Biological Transport/drug effects , Body Weight/drug effects , Cholesterol/blood , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat , Glucose/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin Receptor Substrate Proteins/metabolism , Male , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Rats
8.
Exp Mol Med ; 48: e234, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27197753

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a critical regulator of carbohydrate and lipid metabolism, adipocyte differentiation and inflammatory response. Post-translational modification of PPARγ and its degradation involve several pathways, including the ubiquitin-proteasome system. Here, we identified F-box only protein 9 (FBXO9) as an E3 ubiquitin ligase of PPARγ. We screened interacting partners of PPARγ using immunoprecipitation and mass spectrometric analysis and identified FBXO9 as an E3 ubiquitin ligase of PPARγ. FBXO9 directly interacted with PPARγ through the activation function-1 domain and ligand-binding domain. FBXO9 decreased the protein stability of PPARγ through induction of ubiquitination. We found that the F-box motif of FBXO9 was required for its ubiquitination function. The activity of PPARγ was significantly decreased by FBXO9 overexpression. Furthermore, FBXO9 overexpression in 3T3-L1 adipocytes resulted in decreased levels of endogenous PPARγ and suppression of adipogenesis. These results suggest that FBXO9 is an important enzyme that regulates the stability and activity of PPARγ through ubiquitination.


Subject(s)
Adipocytes/metabolism , F-Box Proteins/metabolism , PPAR gamma/metabolism , Protein Interaction Maps , Ubiquitin-Protein Ligases/metabolism , 3T3-L1 Cells , Animals , Mice , Ubiquitination
9.
Mol Cells ; 38(4): 356-61, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25728751

ABSTRACT

Mitochondrial dysfunction is associated with insulin resistance and diabetes. We previously showed that retinoid X receptor α (RXRα) played an important role in transcriptional regulation of oxidative phosphorylation (OXPHOS) genes in cells with mitochondrial dysfunction caused by mitochondrial DNA mutation. In this study, we investigated whether mitochondrial dysfunction induced by incubation with OXPHOS inhibitors affects insulin receptor substrate 1 (IRS1) mRNA and protein levels and whether RXRα activation or overexpression can restore IRS1 expression. Both IRS1 and RXRα protein levels were significantly reduced when C2C12 myotubes were treated with the OXPHOS complex inhibitors, rotenone and antimycin A. The addition of RXRα agonists, 9-cis retinoic acid (9cRA) and LG1506, increased IRS1 transcription and protein levels and restored mitochondrial function, which ultimately improved insulin signaling. RXRα overexpression also increased IRS1 transcription and mitochondrial function. Because RXRα overexpression, knock-down, or activation by LG1506 regulated IRS1 transcription mostly independently of mitochondrial function, it is likely that RXRα directly regulates IRS1 transcription. Consistent with the hypothesis, we showed that RXRα bound to the IRS1 promoter as a heterodimer with peroxisome proliferator-activated receptor δ (PPARδ). These results suggest that RXRα overexpression or activation alleviates insulin resistance by increasing IRS1 expression.


Subject(s)
Insulin Receptor Substrate Proteins/genetics , Insulin Resistance , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Retinoid X Receptor alpha/biosynthesis , Animals , COS Cells , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Mice , Muscle Fibers, Skeletal/metabolism , Retinoid X Receptor alpha/genetics , Signal Transduction , Transfection
10.
Diabetes ; 64(7): 2420-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25784542

ABSTRACT

Small ubiquitin-like modifier (SUMO)-specific proteases (SENPs) that reverse protein modification by SUMO are involved in the control of numerous cellular processes, including transcription, cell division, and cancer development. However, the physiological function of SENPs in energy metabolism remains unclear. Here, we investigated the role of SENP2 in fatty acid metabolism in C2C12 myotubes and in vivo. In C2C12 myotubes, treatment with saturated fatty acids, like palmitate, led to nuclear factor-κB-mediated increase in the expression of SENP2. This increase promoted the recruitment of peroxisome proliferator-activated receptor (PPAR)δ and PPARγ, through desumoylation of PPARs, to the promoters of the genes involved in fatty acid oxidation (FAO), such as carnitine-palmitoyl transferase-1 (CPT1b) and long-chain acyl-CoA synthetase 1 (ACSL1). In addition, SENP2 overexpression substantially increased FAO in C2C12 myotubes. Consistent with the cell culture system, muscle-specific SENP2 overexpression led to a marked increase in the mRNA levels of CPT1b and ACSL1 and thereby in FAO in the skeletal muscle, which ultimately alleviated high-fat diet-induced obesity and insulin resistance. Collectively, these data identify SENP2 as an important regulator of fatty acid metabolism in skeletal muscle and further implicate that muscle SENP2 could be a novel therapeutic target for the treatment of obesity-linked metabolic disorders.


Subject(s)
Cysteine Endopeptidases/physiology , Fatty Acids/metabolism , Muscle, Skeletal/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Cells, Cultured , Coenzyme A Ligases/genetics , Cysteine Endopeptidases/genetics , Humans , Insulin Resistance , Mice , Muscle Fibers, Skeletal/metabolism , NF-kappa B/physiology , Oxidation-Reduction , PPAR delta/physiology , PPAR gamma/physiology , Palmitic Acid/pharmacology
11.
Rapid Commun Mass Spectrom ; 28(7): 773-80, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24573808

ABSTRACT

RATIONALE: Although in silico prediction of selected reaction monitoring (SRM) peptide transitions is the most commonly used approach in quantitative proteomics, systematically detectable peptide transitions selected from actual experimental data are desirable. Here, we demonstrated the use of two triple quadrupole mass spectrometry (QqQ-MS) operation modes to identify reliable SRM peptide transitions of target peptides selected from a shotgun proteomic linear ion-trap mass spectrometry (LIT-MS) profiling dataset. METHODS: Transition ions (Q1 and Q3 ions) of target peptides were selected from the LIT MS/MS spectra. We performed multiplexed SRM blindly for the selected transition ions of target peptides using QqQ-MS and selected peptide transitions for which the chromatographically aligned and correlated ion intensities to the corresponding fragment ions appeared in the LIT MS/MS spectra. The identities of the peptides were further confirmed by MS/MS spectra acquired via SRM-triggered MS/MS on QqQ-MS. RESULTS: Despite the different MS platforms, we observed similar MS/MS patterns and relative ion abundance using both LIT-MS and QqQ-MS. Therefore, we were able to determine peptide transitions based on matching the chromatographic peak areas of all the selected Q3 ions of target peptides by the order of the corresponding ion intensities in the LIT MS/MS spectra. This approach demonstrated an efficient method to determine SRM peptide transitions, particularly when the target proteins are in low abundance and are therefore not easily detected by the QqQ full MS/MS scan mode. We employed this approach to determine the SRM peptide transitions of mitochondrial oxidative phosphorylation (OXPHOS) proteins involved in mitochondrial ATP synthesis. CONCLUSIONS: The multiplexed product-ion scan mode using QqQ-MS generates systematically detectable peptide transitions in a single liquid chromatography/MS run, in which we were able to identify SRM peptides that represent known target proteins in complex biological samples. The method presented here is easy to implement and has high-throughput capabilities as a result of the short analysis time. It is therefore well suited for the design of optimal SRM experiments.


Subject(s)
Computer Simulation , Ions/chemistry , Peptide Fragments/chemistry , Peptide Mapping/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Humans , Ions/analysis , Molecular Sequence Data , Peptide Fragments/analysis
12.
J Korean Med Sci ; 27(6): 644-52, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22690096

ABSTRACT

Bisphenol A (BPA) has been reported to possess hepatic toxicity. We investigated the hypothesis that BPA, below the no observed adverse effect level (NOAEL), can induce hepatic damage and mitochondrial dysfunction by increasing oxidative stress in the liver. Two doses of BPA, 0.05 and 1.2 mg/kg body weight/day, were administered intraperitoneally for 5 days to mice. Both treatments impaired the structure of the hepatic mitochondria, although oxygen consumption rate and expression of the respiratory complex decreased only at the higher dose. The hepatic levels of malondialdehyde (MDA), a naturally occurring product of lipid peroxidation, increased, while the expression of glutathione peroxidase 3 (GPx3) decreased, after BPA treatment. The expression levels of proinflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) also increased. In HepG2 cells, 10 or 100 nM of BPA also decreased the oxygen consumption rate, ATP production, and the mitochondrial membrane potential. In conclusion, doses of BPA below the NOAEL induce mitochondrial dysfunction in the liver, and this is associated with an increase in oxidative stress and inflammation.


Subject(s)
Liver/drug effects , Mitochondria/metabolism , Phenols/toxicity , Adenosine Triphosphate/metabolism , Animals , Benzhydryl Compounds , Glutathione Peroxidase/metabolism , Hep G2 Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Injections, Intraperitoneal , Interleukin-6/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Tumor Necrosis Factor-alpha/metabolism
13.
Biol Pharm Bull ; 32(12): 2057-60, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952428

ABSTRACT

Hypoxia, which is intimately associated with the biology of breast carcinomas, modulates the level of estrogen receptor (ER) alpha expression and transactivation. We investigated the effect of blocking ER degradation on ERalpha-mediated transactivation under hypoxic conditions using the proteasome inhibitor MG132. Pretreatment with MG132 blocked hypoxia-induced degradation of ERalpha protein. Our data imply that ERalpha proteasomal inhibition is linked to receptor transactivation under hypoxia.


Subject(s)
Estrogen Receptor alpha/metabolism , Hypoxia/metabolism , Leupeptins/pharmacology , Proteasome Inhibitors , Transcriptional Activation , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...