Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33374898

ABSTRACT

Mitochondria are double membrane-bound organelles which are essential for the viability of eukaryotic cells, because they play a crucial role in bioenergetics, metabolism and signaling [...].


Subject(s)
Energy Metabolism/physiology , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Turnover/physiology , Animals , DNA, Mitochondrial/genetics , Energy Metabolism/genetics , Humans , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mitochondrial Turnover/genetics
4.
Diabetes ; 64(9): 3135-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25972572

ABSTRACT

Low mitochondrial number and activity have been suggested as underlying factors in obesity, type 2 diabetes, and metabolic syndrome. However, the stage at which mitochondrial dysfunction manifests in adipose tissue after the onset of obesity remains unknown. Here we examined subcutaneous adipose tissue (SAT) samples from healthy monozygotic twin pairs, 22.8-36.2 years of age, who were discordant (ΔBMI >3 kg/m(2), mean length of discordance 6.3 ± 0.3 years, n = 26) and concordant (ΔBMI <3 kg/m(2), n = 14) for body weight, and assessed their detailed mitochondrial metabolic characteristics: mitochondrial-related transcriptomes with dysregulated pathways, mitochondrial DNA (mtDNA) amount, mtDNA-encoded transcripts, and mitochondrial oxidative phosphorylation (OXPHOS) protein levels. We report global expressional downregulation of mitochondrial oxidative pathways with concomitant downregulation of mtDNA amount, mtDNA-dependent translation system, and protein levels of the OXPHOS machinery in the obese compared with the lean co-twins. Pathway analysis indicated downshifting of fatty acid oxidation, ketone body production and breakdown, and the tricarboxylic acid cycle, which inversely correlated with adiposity, insulin resistance, and inflammatory cytokines. Our results suggest that mitochondrial biogenesis, oxidative metabolic pathways, and OXPHOS proteins in SAT are downregulated in acquired obesity, and are associated with metabolic disturbances already at the preclinical stage.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Turnover/genetics , Obesity/genetics , Subcutaneous Fat/metabolism , Twins, Monozygotic , Adult , Case-Control Studies , Citric Acid Cycle/genetics , Cytokines/immunology , Cytokines/metabolism , DNA, Mitochondrial/metabolism , Fatty Acids/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Inflammation , Insulin Resistance/genetics , Ketone Bodies/metabolism , Male , Mitochondria/metabolism , Obesity/immunology , Obesity/metabolism , Oxidative Phosphorylation , Subcutaneous Fat/immunology
5.
J Cell Physiol ; 230(10): 2330-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25736533

ABSTRACT

The microphthalmia (MiT) family of transcription factors is an important mediator of metabolism. Family members Mitf and Tfeb directly regulate the expression of the master regulator of metabolism, peroxisome-proliferator activated receptor gamma coactivator-1 alpha (Pgc-1alpha), in melanomas and in the liver, respectively. Pgc-1alpha is enriched in tissues with high oxidative capacity and plays an important role in the regulation of mitochondrial biogenesis and cellular metabolism. In skeletal muscle, Pgc-1alpha affects many aspects of muscle functionally such as endurance, fiber-type switching, and insulin sensitivity. Tfe3 also regulates muscle metabolic genes that enhance insulin sensitivity in skeletal muscle. Tfe3 has not yet been shown to regulate Pgc-1alpha expression. Our results reported here show that Tfe3 directly regulates Pgc-1alpha expression in myotubes. Tfe3 ectopic expression induces Pgc-1alpha, and Tfe3 silencing suppresses Pgc-1alpha expression. This regulation is direct, as shown by Tfe3's binding to E-boxes on the Pgc-1alpha proximal promoter. We conclude that Tfe3 is a critical transcription factor that regulates Pgc-1alpha gene expression in myotubes. Since Pgc-1alpha coactivates numerous biological programs in diverse tissues, the regulation of its expression by upstream transcription factors such Tfe3 implies potential opportunities for the treatment of diseases where modulation of Pgc-1alpha expression may have important clinical outcomes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Mitochondrial Turnover/genetics , Muscle, Skeletal/metabolism , Transcription Factors/metabolism , Animals , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics
6.
Neurobiol Aging ; 36(3): 1282-92, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582749

ABSTRACT

Mitochondrial dysfunction, especially a defect in mitochondrial biogenesis, is an early and prominent feature of Alzheimer's disease (AD). Previous studies demonstrated that the number of mitochondria is significantly reduced in susceptible hippocampal neurons from AD patients. Neural stem cell (NSC) transplantation in AD-like mice can compensate for the neuronal loss resulting from amyloid-beta protein deposition. The effects of NSC transplantation on mitochondrial biogenesis and cognitive function in AD-like mice, however, are poorly understood. In this study, we injected NSCs or vehicle into 12-month-old amyloid precursor protein (APP)/PS1 transgenic mice, a mouse model of AD-like pathology. The effects of NSC transplantation on cognitive function, the amount of mitochondrial DNA, the expression of mitochondrial biogenesis factors and mitochondria-related proteins, and mitochondrial morphology were investigated. Our results show that in NSC-injected APP/PS1 (Tg-NSC) mice, the cognitive function, number of mitochondria, and expression of mitochondria-related proteins, specifically the mitochondrial fission factors (dynamin-related protein 1 [Drp1] and fission 1 [Fis1]) and the mitochondrial fusion factor optic atrophy 1 (OPA1), were significantly increased compared with those in age-matched vehicle-injected APP/PS1 (Tg-Veh) mice, whereas the expression of mitochondrial fusion factors mitofusion 1 (Mfn1) and Mfn2 was significantly decreased. These data indicate that NSC transplantation may enhance mitochondria biogenesis and further rescue cognitive deficits in AD-like mice.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/therapy , Mitochondrial Turnover , Neural Stem Cells/transplantation , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cognition , DNA, Mitochondrial/metabolism , Disease Models, Animal , Dynamins/genetics , Dynamins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Turnover/genetics
7.
Aging (Albany NY) ; 7(1): 38-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25596819

ABSTRACT

To elucidate gene expression pathways underlying age-associated impairment in influenza vaccine response, we screened young (age 21-30) and older (age≥65) adults receiving influenza vaccine in two consecutive seasons and identified those with strong or absent response to vaccine, including a subset of older adults meeting criteria for frailty. PBMCs obtained prior to vaccination (Day 0) and at day 2 or 4, day 7 and day 28 post-vaccine were subjected to gene expression microarray analysis. We defined a response signature and also detected induction of a type I interferon response at day 2 and a plasma cell signature at day 7 post-vaccine in young responders. The response signature was dysregulated in older adults, with the plasma cell signature induced at day 2, and was never induced in frail subjects (who were all non-responders). We also identified a mitochondrial signature in young vaccine responders containing genes mediating mitochondrial biogenesis and oxidative phosphorylation that was consistent in two different vaccine seasons and verified by analyses of mitochondrial content and protein expression. These results represent the first genome-wide transcriptional profiling analysis of age-associated dynamics following influenza vaccination, and implicate changes in mitochondrial biogenesis and function as a critical factor in human vaccine responsiveness.


Subject(s)
Aging/genetics , DNA, Mitochondrial/metabolism , Gene Expression Regulation/drug effects , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Leukocytes, Mononuclear/drug effects , Mitochondria/drug effects , Vaccination , Adult , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Aging/metabolism , Cells, Cultured , Female , Gene Expression Profiling/methods , Genome-Wide Association Study , Humans , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mitochondria/immunology , Mitochondria/metabolism , Mitochondrial Turnover/drug effects , Mitochondrial Turnover/genetics , Oligonucleotide Array Sequence Analysis , Oxidative Phosphorylation/drug effects , Seasons , Time Factors , Treatment Outcome , Young Adult
8.
FEBS J ; 282(4): 647-72, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25495651

ABSTRACT

Members of the PGC-1 family of coactivators have been revealed as key players in the regulation of energy metabolism. Early gain- and loss-of-function studies led to the conclusion that all members of the PGC-1 family (PGC-1α, PGC-1ß and PRC) play redundant roles in the control of mitochondrial biogenesis by regulating overlapping gene expression programs. Regardless of this, all PGC-1 coactivators also appeared to differ in the stimuli to which they respond to promote mitochondrial gene expression. Although PGC-1α was found to be induced by different physiological or pharmacological cues, PGC-1ß appeared to be unresponsive to such stimuli. Consequently, it has long been widely accepted that PGC-1α acts as a mediator of mitochondrial biogenesis induced by cues that signal high-energy needs, whereas the role of PGC-1ß is restricted to the maintenance of basal mitochondrial function. By contrast, the function of PRC appears to be restricted to the regulation of gene expression in proliferating cells. However, recent studies using tissue-specific mouse models that lack or overexpress different PGC-1 coactivators have provided emerging evidence not only supporting new roles for PGC-1s, but also redefining some of the paradigms related to the precise function and mode of action of PGC-1 coactivators in mitochondrial biogenesis. The present review discusses some of the new findings regarding the control of mitochondrial gene expression by PGC-1 coactivators in a tissue-specific context, as well as newly-uncovered functions of PGC-1s beyond mitochondrial biogenesis, and their link to pathologies, such as diabetes, muscular dystrophies, neurodegenerative diseases or cancer.


Subject(s)
Energy Metabolism/physiology , Mitochondria/metabolism , Mitochondrial Turnover/physiology , Transcription Factors/metabolism , Animals , Energy Metabolism/genetics , Humans , Mice , Mitochondria/genetics , Mitochondrial Turnover/genetics , Transcription Factors/genetics
9.
Cell Death Dis ; 5: e1515, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25375380

ABSTRACT

Mitochondrial biogenesis and mitophagy are recognized as critical processes underlying mitochondrial homeostasis. However, the molecular pathway(s) coordinating the balance between these cellular programs is still poorly investigated. Here, we show an induction of the nuclear and mitochondrial peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (PGC-1α) during myogenesis, which in turn co-activates the transcription of nuclear and mtDNA-encoded mitochondrial genes. We demonstrate that PGC-1α also buffers oxidative stress occurring during differentiation by promoting the expression of antioxidant enzymes. Indeed, by downregulating PGC-1α, we observed an impairment of antioxidants expression, which was accompanied by a significant reactive oxygen species (ROS) burst and increase of oxidative damage to proteins. In parallel, we detected a decrease of mitochondrial mass and function as well as increased mitophagy through the ROS/FOXO1 pathway. Upon PGC-1α downregulation, we found ROS-dependent nuclear translocation of FOXO1 and transcription of its downstream targets including mitophagic genes such as LC3 and PINK1. Such events were significantly reverted after treatment with the antioxidant Trolox, suggesting that PGC-1α assures mitochondrial integrity by indirectly buffering ROS. Finally, the lack of PGC-1α gave rise to a decrease in MYOG and a strong induction of atrophy-related ubiquitin ligases FBXO32 (FBXO32), indicative of a degenerative process. Overall, our results reveal that in myotubes, PGC-1α takes center place in mitochondrial homeostasis during differentiation because of its ability to avoid ROS-mediated removal of mitochondria.


Subject(s)
Mitochondria/metabolism , Mitochondrial Turnover/genetics , Muscle Cells/metabolism , Muscle Development/genetics , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Animals , Antioxidants/pharmacology , Cell Differentiation , Cell Line , Chromans/pharmacology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitophagy , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myogenin/genetics , Myogenin/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Transcription Factors/metabolism
10.
PLoS One ; 9(11): e112360, 2014.
Article in English | MEDLINE | ID: mdl-25402171

ABSTRACT

Mitochondria play a pivotal role in most eukaryotic cells, as they are responsible for the generation of energy and diverse metabolic intermediates for many cellular events. During endosymbiosis, approximately 99% of the genes encoded by the mitochondrial genome were transferred into the host nucleus, and mitochondria import more than 1000 nuclear-encoded proteins from the cytosol to maintain structural integrity and fundamental functions, including DNA replication, mRNA transcription and RNA metabolism of dozens of mitochondrial genes. In metazoans, a family of nuclear-encoded proteins called the mitochondrial transcription termination factors (mTERFs) regulates mitochondrial transcription, including transcriptional termination and initiation, via their DNA-binding activities, and the dysfunction of individual mTERF members causes severe developmental defects. Arabidopsis thaliana and Oryza sativa contain 35 and 48 mTERFs, respectively, but the biological functions of only a few of these proteins have been explored. Here, we investigated the biological role and molecular mechanism of Arabidopsis mTERF15 in plant organelle metabolism using molecular genetics, cytological and biochemical approaches. The null homozygous T-DNA mutant of mTERF15, mterf15, was found to result in substantial retardation of both vegetative and reproductive development, which was fully complemented by the wild-type genomic sequence. Surprisingly, mitochondria-localized mTERF15 lacks obvious DNA-binding activity but processes mitochondrial nad2 intron 3 splicing through its RNA-binding ability. Impairment of this splicing event not only disrupted mitochondrial structure but also abolished the activity of mitochondrial respiratory chain complex I. These effects are in agreement with the severe phenotype of the mterf15 homozygous mutant. Our study suggests that Arabidopsis mTERF15 functions as a splicing factor for nad2 intron 3 splicing in mitochondria, which is essential for normal plant growth and development.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Electron Transport Complex I/metabolism , Introns , Mitochondria/genetics , Mitochondria/metabolism , RNA Splicing , Arabidopsis/growth & development , Membrane Potential, Mitochondrial , Mitochondria/ultrastructure , Mitochondrial Turnover/genetics , Mutation , Phenotype , Protein Transport
11.
Mar Drugs ; 12(8): 4602-15, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25196935

ABSTRACT

Echinochrome A (Ech A) is a natural pigment from sea urchins that has been reported to have antioxidant properties and a cardio protective effect against ischemia reperfusion injury. In this study, we ascertained whether Ech A enhances the mitochondrial biogenesis and oxidative phosphorylation in rat cardio myoblast H9c2 cells. To study the effects of Ech A on mitochondrial biogenesis, we measured mitochondrial mass, level of oxidative phosphorylation, and mitochondrial biogenesis regulatory gene expression. Ech A treatment did not induce cytotoxicity. However, Ech A treatment enhanced oxygen consumption rate and mitochondrial ATP level. Likewise, Ech A treatment increased mitochondrial contents in H9c2 cells. Furthermore, Ech A treatment up-regulated biogenesis of regulatory transcription genes, including proliferator-activated receptor gamma co-activator (PGC)-1α, estrogen-related receptor (ERR)-α, peroxisome proliferator-activator receptor (PPAR)-γ, and nuclear respiratory factor (NRF)-1 and such mitochondrial transcription regulatory genes as mitochondrial transcriptional factor A (TFAM), mitochondrial transcription factor B2 (TFB2M), mitochondrial DNA direct polymerase (POLMRT), single strand binding protein (SSBP) and Tu translation elongation factor (TUFM). In conclusion, these data suggest that Ech A is a potentiated marine drug which enhances mitochondrial biogenesis.


Subject(s)
Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Turnover/drug effects , Mitochondrial Turnover/genetics , Myoblasts, Cardiac/drug effects , Naphthoquinones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Line , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Myoblasts, Cardiac/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Rats , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
12.
Neuromolecular Med ; 16(4): 714-26, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25081190

ABSTRACT

Origins of onset and progression of motor neurodegeneration in amyotrophic lateral sclerosis (ALS) are not clearly known, but may include impairment of mitochondrial bioenergetics. We used quantitative PCR approaches to analyze the mitochondrial oxidative phosphorylation (OXPHOS) transcriptomes of spinal cord tissue and peripheral blood mononuclear cells (PBMC) from persons with sporadic ALS compared with those without neurological disease. Expression measurements of 88 different nuclear (n) and mitochondrial (mt) DNA-encoded OXPHOS genes showed mtDNA-encoded respiratory gene expression was significantly decreased in ALS spinal cord by 78-84% (ANOVA p < 0.002). We observed the same phenomenon in freshly isolated PBMC from ALS patients (reduced 24-35%, ANOVA p < 0.001) and reproduced it in a human neural stem cell model treated with 2',3'-dideoxycytidine (ddC) (reduced 52-78%, ANOVA p < 0.001). nDNA-encoded OXPHOS genes showed heterogeneously and mostly decreased expression in ALS spinal cord tissue. In contrast, ALS PBMC and ddC-treated stem cells showed no significant change in expression of nDNA OXPHOS genes compared with controls. Genes related to mitochondrial biogenesis (PGC-1α, TFAM, ERRα, NRF1, NRF2 and POLG) were queried with inconclusive results. Here, we demonstrate there is a systemic decrease in mtDNA gene expression in ALS central and peripheral tissues that support pursuit of bioenergetic-enhancing therapies. We also identified a combined nDNA and mtDNA gene set (n = 26), downregulated in spinal cord tissue that may be useful as a biomarker in the development of cell-based ALS models.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA, Mitochondrial/genetics , Gene Expression Regulation , Genes, Mitochondrial , Leukocytes, Mononuclear/metabolism , Mitochondria/physiology , Oxidative Phosphorylation , RNA, Messenger/biosynthesis , Spinal Cord/metabolism , Transcriptome , Aged , Amyotrophic Lateral Sclerosis/metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Mitochondrial Turnover/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , RNA, Messenger/genetics , Transcription Factors/physiology , Zalcitabine/pharmacology
13.
Hum Mol Genet ; 23(24): 6528-41, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25027327

ABSTRACT

Brain bioenergetic function declines in some neurodegenerative diseases, this may influence other pathologies and administering bioenergetic intermediates could have therapeutic value. To test how one intermediate, oxaloacetate (OAA) affects brain bioenergetics, insulin signaling, inflammation and neurogenesis, we administered intraperitoneal OAA, 1-2 g/kg once per day for 1-2 weeks, to C57Bl/6 mice. OAA altered levels, distributions or post-translational modifications of mRNA and proteins (proliferator-activated receptor-gamma coactivator 1α, PGC1 related co-activator, nuclear respiratory factor 1, transcription factor A of the mitochondria, cytochrome oxidase subunit 4 isoform 1, cAMP-response element binding, p38 MAPK and adenosine monophosphate-activated protein kinase) in ways that should promote mitochondrial biogenesis. OAA increased Akt, mammalian target of rapamycin and P70S6K phosphorylation. OAA lowered nuclear factor κB nucleus-to-cytoplasm ratios and CCL11 mRNA. Hippocampal vascular endothelial growth factor mRNA, doublecortin mRNA, doublecortin protein, doublecortin-positive neuron counts and neurite length increased in OAA-treated mice. (1)H-MRS showed OAA increased brain lactate, GABA and glutathione thereby demonstrating metabolic changes are detectable in vivo. In mice, OAA promotes brain mitochondrial biogenesis, activates the insulin signaling pathway, reduces neuroinflammation and activates hippocampal neurogenesis.


Subject(s)
Hippocampus/drug effects , Insulin/metabolism , Mitochondrial Turnover/drug effects , Neurogenesis/drug effects , Oxaloacetic Acid/administration & dosage , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Doublecortin Domain Proteins , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Expression Regulation , Glutathione/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Inflammation/prevention & control , Injections, Intraperitoneal , Insulin/genetics , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Turnover/genetics , Neurogenesis/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Diabetologia ; 57(9): 1957-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24973963

ABSTRACT

AIMS/HYPOTHESIS: Adipose tissue from obese and insulin-resistant individuals showed altered expression of several iron-related genes in a recent study, suggesting that iron might have an important role in adipogenesis. To investigate this possible role, we aimed to characterise the effects of iron on adipocyte differentiation. METHODS: Intracellular iron deficiency was achieved using two independent approaches: deferoxamine administration (20 and 100 µmol/l) and transferrin knockdown (TF KD). The effects of added FeSO4, holo-transferrin and palmitate were studied during human and 3T3-L1 adipocyte differentiation. Finally, the relationship between iron-related and mitochondrial-related genes was investigated in human adipose tissue. RESULTS: Most adipose tissue iron-related genes were predominantly expressed in adipocytes compared with stromal vascular cells. Of note, transferrin gene and protein expression increased significantly during adipocyte differentiation. Both deferoxamine and TF KD severely blunted adipocyte differentiation in parallel with increased inflammatory mRNAs. These effects were reversed in a dose-dependent manner after iron supplementation. Palmitate administration also led to a state of functional intracellular iron deficiency, with decreased Tf gene expression and iron uptake during adipocyte differentiation, which was reversed with transferrin co-treatment. On the other hand, iron in excess impaired differentiation, but this antiadipogenic effect was less pronounced than under iron chelation. Of interest, expression of several genes involved in mitochondrial biogenesis occurred in parallel with expression of iron-related genes both during adipogenesis and in human adipose tissue. CONCLUSIONS/INTERPRETATION: Precise and fine-tuned iron availability is essential to achieve optimal adipocyte differentiation, possibly modulating adipocyte mitochondrial biogenesis.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , Mitochondrial Turnover/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice , Mitochondrial Turnover/genetics
15.
BMC Plant Biol ; 14: 144, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24886177

ABSTRACT

BACKGROUND: Uncoupling protein one (UCP1) is a mitochondrial inner membrane protein capable of uncoupling the electrochemical gradient from adenosine-5'-triphosphate (ATP) synthesis, dissipating energy as heat. UCP1 plays a central role in nonshivering thermogenesis in the brown adipose tissue (BAT) of hibernating animals and small rodents. A UCP1 ortholog also occurs in plants, and aside from its role in uncoupling respiration from ATP synthesis, thereby wasting energy, it plays a beneficial role in the plant response to several abiotic stresses, possibly by decreasing the production of reactive oxygen species (ROS) and regulating cellular redox homeostasis. However, the molecular mechanisms by which UCP1 is associated with stress tolerance remain unknown. RESULTS: Here, we report that the overexpression of UCP1 increases mitochondrial biogenesis, increases the uncoupled respiration of isolated mitochondria, and decreases cellular ATP concentration. We observed that the overexpression of UCP1 alters mitochondrial bioenergetics and modulates mitochondrial-nuclear communication, inducing the upregulation of hundreds of nuclear- and mitochondrial-encoded mitochondrial proteins. Electron microscopy analysis showed that these metabolic changes were associated with alterations in mitochondrial number, area and morphology. Surprisingly, UCP1 overexpression also induces the upregulation of hundreds of stress-responsive genes, including some involved in the antioxidant defense system, such as superoxide dismutase (SOD), glutathione peroxidase (GPX) and glutathione-S-transferase (GST). As a consequence of the increased UCP1 activity and increased expression of oxidative stress-responsive genes, the UCP1-overexpressing plants showed reduced ROS accumulation. These beneficial metabolic effects may be responsible for the better performance of UCP1-overexpressing lines in low pH, high salt, high osmolarity, low temperature, and oxidative stress conditions. CONCLUSIONS: Overexpression of UCP1 in the mitochondrial inner membrane induced increased uncoupling respiration, decreased ROS accumulation under abiotic stresses, and diminished cellular ATP content. These events may have triggered the expression of mitochondrial and stress-responsive genes in a coordinated manner. Because these metabolic alterations did not impair plant growth and development, UCP1 overexpression can potentially be used to create crops better adapted to abiotic stress conditions.


Subject(s)
Arabidopsis/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Turnover/genetics , Nicotiana/genetics , Nicotiana/physiology , Stress, Physiological/genetics , Adenosine Triphosphate/metabolism , Cell Respiration/genetics , Gene Expression Regulation, Plant , Genes, Plant , Mitochondria/metabolism , Mitochondria/ultrastructure , Phenotype , Plant Leaves/metabolism , Plant Leaves/ultrastructure , Plants, Genetically Modified , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Transgenes , Uncoupling Protein 1 , Up-Regulation/genetics
16.
Diabetes ; 63(11): 3734-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24947361

ABSTRACT

Adipocytes are the primary cells in the body that store excess energy as triglycerides. To perform this specialized function, adipocytes rely on their mitochondria; however, the role of adipocyte mitochondria in the regulation of adipose tissue homeostasis and its impact on metabolic regulation is not understood. We developed a transgenic mouse model, Mito-Ob, overexpressing prohibitin (PHB) in adipocytes. Mito-Ob mice developed obesity due to upregulation of mitochondrial biogenesis in adipocytes. Of note, Mito-Ob female mice developed more visceral fat than male mice. However, female mice exhibited no change in glucose homeostasis and had normal insulin and high adiponectin levels, whereas male mice had impaired glucose homeostasis, compromised brown adipose tissue structure, and high insulin and low adiponectin levels. Mechanistically, we found that PHB overexpression enhances the cross talk between the mitochondria and the nucleus and facilitates mitochondrial biogenesis. The data suggest a critical role of PHB and adipocyte mitochondria in adipose tissue homeostasis and reveal sex differences in the effect of PHB-induced adipocyte mitochondrial remodeling on whole-body metabolism. Targeting adipocyte mitochondria may provide new therapeutic opportunities for the treatment of obesity, a major risk factor for type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Mitochondrial Turnover/physiology , Repressor Proteins/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Glucose/metabolism , Homeostasis , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, Transgenic , Mitochondrial Turnover/genetics , Obesity/genetics , Obesity/metabolism , Prohibitins , Repressor Proteins/genetics , Sex Factors
17.
Int J Biochem Cell Biol ; 53: 9-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24792670

ABSTRACT

Delphinidin, an anthocyanin present in red wine, has been reported to exert vasculoprotective properties on endothelial cells, including vasorelaxing and anti-apoptotic effects. Moreover, delphinidin treatment in a rat model of post-ischemic neovascularization has been described to exert anti-angiogenic property. Angiogenesis is an energetic process and VEGF-induced angiogenesis is associated with mitochondrial biogenesis. However, whether delphinidin induces changes in mitochondrial biogenesis has never been addressed. Effects of delphinidin were investigated in human endothelial cells at a concentration described to be anti-angiogenic in vitro (10(-2)g/l). mRNA expression of mitochondrial biogenesis factors, mitochondrial respiration, DNA content and enzyme activities were assessed after 48 h of stimulation. Delphinidin increased mRNA expression of several mitochondrial biogenesis factors, including NRF1, ERRα, Tfam, Tfb2m and PolG but did not affect neither mitochondrial respiration, DNA content nor enzyme activities. In presence of delphinidin, VEGF failed to increase mitochondrial respiration, DNA content, complex IV activity and Akt activation in endothelial cells. These results suggest a possible association between inhibition of VEGF-induced mitochondrial biogenesis through Akt pathway by delphinidin and its anti-angiogenic effect, providing a novel mechanism sustaining the beneficial effect of delphinidin against pathologies associated with excessive angiogenesis such as cancers.


Subject(s)
Anthocyanins/administration & dosage , Cell Respiration/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Angiogenesis Inhibitors/administration & dosage , Animals , Apoptosis/drug effects , Cell Respiration/drug effects , Endothelial Cells/drug effects , Humans , Mitochondrial Turnover/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects
18.
Article in English | MEDLINE | ID: mdl-24789872

ABSTRACT

Mitochondria, the powerhouses of the cell, face two imperatives concerning biogenesis. The first is the requirement for dividing cells to replicate their mitochondrial content by growth of existing mitochondria. The second is the dynamic regulation of mitochondrial content in response to organismal and cellular cues (e.g., exercise, caloric restriction, energy status, temperature). MYC provides the clearest example of a programmed expansion of mitochondrial content linked to the cell cycle. As an oncogene, MYC also presents intriguing questions about the role of its mitochondrial targets in cancer-related phenotypes, such as the Warburg effect and MYC-dependent apoptosis.


Subject(s)
Genes, myc/physiology , Mitochondria/genetics , Mitochondrial Turnover/genetics , Cell Proliferation/genetics , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcriptome/genetics
19.
Exp Gerontol ; 56: 59-68, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24699405

ABSTRACT

Extrinsic skin ageing converges on the dermis, a post-mitotic tissue compartment consisting of extracellular matrix and long-lived fibroblasts prone to damage accumulation and maladaptation. Aged human fibroblasts exhibit mitochondrial and nuclear dysfunctions, which may be a cause or consequence of ageing. We report on a systematic study of human dermal fibroblasts retrieved from female donors aged 20-67 years and analysed ex vivo at low population doubling precluding replicative senescence. According to gene set enrichment analysis of genome wide array data, the most prominent age-associated change of the transcriptome was decreased expression of mitochondrial genes. Consistent with that, mitochondrial content and cell proliferation declined with donor age. This was associated with upregulation of AMP-dependent protein kinase (AMPK), increased mRNA levels of PPARγ-coactivator 1α (PGC1A) and decreased levels of NAD(+)-dependent deacetylase sirtuin 1. In the old cells the PGC1A-mediated mito-biogenetic response to direct AMPK-stimulation by AICAR was undiminished, while the PGC1A-independent mito-biogenetic response to starvation was attenuated and accompanied by increased ROS-production. In summary, these observations suggest an age-associated decline in PGC1A-independent mito-biogenesis, which is insufficiently compensated by upregulation of the AMPK/PGC1A-axis leading under baseline conditions to decreased mitochondrial content and reductive overload of residual respiratory capacity.


Subject(s)
Aging/metabolism , Energy Metabolism , Fibroblasts/metabolism , Mitochondria/metabolism , Mitochondrial Turnover , Skin Aging , Skin/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Adult , Age Factors , Aged , Aging/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Cell Proliferation , Cells, Cultured , Cellular Senescence , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Fibroblasts/drug effects , Gene Expression Regulation , Humans , Middle Aged , Mitochondria/drug effects , Mitochondrial Turnover/drug effects , Mitochondrial Turnover/genetics , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Ribonucleotides/pharmacology , Signal Transduction , Sirtuin 1/metabolism , Skin/drug effects , Skin Aging/genetics , Transcription Factors/genetics , Young Adult
20.
Cell Cycle ; 13(6): 890-7, 2014.
Article in English | MEDLINE | ID: mdl-24552811

ABSTRACT

The growing epidemic of type 2 diabetes mellitus (T2DM) and obesity is largely attributed to the current lifestyle of over-consumption and physical inactivity. As the primary platform controlling metabolic and energy homeostasis, mitochondria show aberrant changes in T2DM and obese subjects. While the underlying mechanism is under extensive investigation, epigenetic regulation is now emerging to play an important role in mitochondrial biogenesis, function, and dynamics. In line with lifestyle modifications preventing mitochondrial alterations and metabolic disorders, exercise has been shown to change DNA methylation of the promoter of PGC1α to favor gene expression responsible for mitochondrial biogenesis and function. In this article we discuss the epigenetic mechanism of mitochondrial alteration in T2DM and obesity, and the effects of lifestyle on epigenetic regulation. Future studies designed to further explore and integrate the epigenetic mechanisms with lifestyle modification may lead to interdisciplinary interventions and novel preventive options for mitochondrial alteration and metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Mitochondria/metabolism , Obesity/metabolism , Animals , Energy Metabolism , Humans , Life Style , Mitochondria/genetics , Mitochondrial Turnover/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...