Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
3.
J Sport Health Sci ; 7(2): 191-196, 2018 Apr.
Article in English | MEDLINE | ID: mdl-30356443

ABSTRACT

BACKGROUND: Exercise promotes numerous phenotypic adaptations in skeletal muscle that contribute to improved function and metabolic capacity. An emerging body of evidence suggests that skeletal muscle also releases a myriad of factors during exercise, termed "myokines". The purpose of this study was to examine the effects of high-intensity interval training (HIIT) on the acute regulation of the mRNA expression of several myokines, including the prototypical myokine interleukin-6 (IL-6), and recently identified myokines fibronectin type III domain-containing protein 5 (FNDC5) (irisin) and meteorin-like protein (METRNL). METHODS: Both before and after a 20-day period of twice-daily high-volume HIIT, 9 healthy males (20.5 ± 1.5 years performed a standardized bout of high-intensity interval exercise (HIIE; 5 × 4 min at ~80% pretraining peak power output) with skeletal muscle biopsy samples (vastus lateralis) obtained at rest, immediately following exercise, and at 3 h recovery. RESULTS: Before training, a single bout of HIIE increased IL-6 (p < 0.05) and METRNL (p < 0.05) mRNA expression measured at 3 h recovery when compared to rest. Following 20 days of HIIT, IL-6 and FNDC5 mRNA were increased at 3 h recovery from the standardized HIIE bout when compared to rest (both p < 0.05). Resting METRNL and FNDC5 mRNA expression were higher following training (p < 0.05), and there was an overall increase in FNDC5 mRNA post-training (main effect of training, p < 0.05). CONCLUSION: In human skeletal muscle (1) an acute bout of HIIE can induce upregulation of skeletal muscle IL-6 mRNA both before and after a period of intensified HIIT; (2) Resting and overall FNDC5 mRNA expression is increased by 20 days of HIIT; and (3) METRNL mRNA expression is responsive to both acute HIIE and short-term intense HIIT. Future studies are needed to confirm these findings at the protein and secretion level in humans.

6.
Article in English | MEDLINE | ID: mdl-28490541

ABSTRACT

Habitual endurance exercise training is associated with multisystemic metabolic adaptations that lower the risk of inactivity-associated disorders such as obesity and type 2 diabetes mellitus (T2DM). Identification of complex systemic signaling networks responsible for these benefits are of great interest because of their therapeutic potential in metabolic diseases; however, specific signals that modulate the multisystemic benefits of exercise in multiple tissues and organs are only recently being discovered. Accumulated evidence suggests that muscle and other tissues have an endocrine function and release peptides and nucleic acids into the circulation in response to acute endurance exercise to mediate the multisystemic adaptations. Factors released from skeletal muscle have been termed myokines and we propose that the total of all factors released in response to endurance exercise (including peptides, nucleic acids, and metabolites) be termed, "exerkines." We propose that many of the exerkines are released within extracellular vesicles called exosomes, which regulate peripheral organ cross talk. Exosomes (30-140 nm) and larger microvesicles [MVs] (100-1000 nm) are subcategories of extracellular vesicles that are released into the circulation. Exosomes contain peptides and several nucleic acids (microRNA [miRNA], messenger RNA [mRNA], mitochondrial DNA [mtDNA]) and are involved in intercellular/tissue exchange of their contents. An acute bout of endurance exercise increases circulating exosomes that are hypothesized to mediate organ cross talk to promote systemic adaptation to endurance exercise. Further support for the role of exosomes (and possibly MVs) in mediating the systemic benefits of exercise comes from the fact that the majority of the previously reported myokines/exerkines are found in extracellular vesicles databases (Vesiclepedia and ExoCarta). We propose that exosomes isolated from athletes following exercise or exosomes bioengineered to incorporate one or many of known exerkines will be therapeutically useful in the treatment of obesity, T2DM, and other aging-associated metabolic disorders.


Subject(s)
Exercise/physiology , Exosomes/metabolism , Exosomes/transplantation , Metabolic Diseases/metabolism , Metabolic Diseases/therapy , Adaptation, Physiological , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Extracellular Vesicles/metabolism , Humans , Muscle, Skeletal/metabolism , Obesity/metabolism , Obesity/therapy , Physical Endurance
7.
Eur J Hum Genet ; 25(6): 744-751, 2017 06.
Article in English | MEDLINE | ID: mdl-28295037

ABSTRACT

Recent research has suggested that transmembrane protein 65 (TMEM65) is localized within the inner mitochondrial membrane. Little else is known about its function. In this study we investigated the location and function of TMEM65. Further, we report the functional consequences of a novel homozygous splice variant (c.472+1G>A) in the TMEM65 gene in a patient with mitochondrial encephalomyopathy. Here we investigated the location of TMEM65 by immunofluorescence staining of the protein and by immunoblotting of the isolated mitochondrial fractions in healthy fibroblasts and those from the patient. To study the function of TMEM65 we knocked down mRNA using TMEM65-specific siRNA, and measured mitochondrial function by enzymology, protein abundance and oxygen consumption rate in fibroblasts. Subcellular fractionation confirmed that the TMEM65 protein was present in the inner mitochondrial membrane. Knocking down TMEM65 expression in dermal fibroblasts severely affected mitochondrial content and respiration rate. Further evidence for the essential role of TMEM65 in mitochondrial function came from the demonstration of severe cellular and clinical consequences resulting from the novel TMEM65 gene mutation. In conclusion, these findings suggest that TMEM65, an inner mitochondrial membrane protein, plays a significant role in mitochondrial respiratory chain function. We also provide the first evidence that a mutation in the TMEM65 gene results in mitochondrial dysfunction and a severe mitochondrial encephalomyopathy phenotype.


Subject(s)
Membrane Proteins/genetics , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/genetics , Mutation , Cell Respiration , Cells, Cultured , Child, Preschool , Female , Fibroblasts/metabolism , Humans , Membrane Proteins/metabolism , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Oxygen/metabolism , RNA Splicing
8.
Exp Physiol ; 102(3): 354-365, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28118678

ABSTRACT

NEW FINDINGS: What is the central question of this study? Are there sex-based differences in the acute skeletal muscle response to sprint interval training (SIT)? What is the main finding and its importance? In response to a SIT protocol that involved three 20 s bouts of 'all-out' cycling, the expression of multiple genes associated with mitochondrial biogenesis, metabolic control and structural remodelling was largely similar between men and women matched for fitness. Our findings cannot explain previous reports of sex-based differences in the adaptive response to SIT and suggest that the mechanistic basis for these differences remains to be elucidated. A few studies have reported sex-based differences in response to several weeks of sprint interval training (SIT). These findings may relate to sex-specific responses to an acute session of SIT. We tested the hypothesis that the acute skeletal muscle response to SIT differs between sexes. Sedentary but healthy men (n = 10) and women (n = 9) were matched for age (22 ± 3 versus 22 ± 3 years old) and cardiorespiratory fitness [45 ± 7 versus 43 ± 10 ml O2  (kg fat-free mass)-1  min-1 ], with women tested in the mid-follicular phase of their menstrual cycles. Subjects performed three 20 s 'all-out' cycling efforts against a resistance of 5% of body mass, interspersed with 2 min of recovery. Relative mean power outputs [7.6 ± 0.5 versus 7.5 ± 0.9 W (kg fat-free mass)-1 ] were similar between men and women (P > 0.05). Furthermore, there were no differences in the exercise-induced changes in mRNA expression of PGC-1α, PRC, PPARD, SIRT1, RIP140, HSL, HKII, PDK4, PDP1, FOXO3, MURF-1, Myf5, MyoD and VEGFA at 3 h of recovery versus rest (P < 0.05, main effect of time). The only sex-specific responses to exercise were an increase in the mRNA expression of GLUT4 and LPL in women only and Atrogin-1 in men only (P < 0.05). Women also had higher expression of HKII and lower expression of FOXO3 compared with men (P < 0.05, main effect of sex). We conclude that the acute skeletal muscle response to SIT is largely similar in young men and women. The mechanistic basis for sex-based differences in response to several weeks of SIT that has been previously reported remains to be elucidated.


Subject(s)
Exercise/physiology , Muscle, Skeletal/metabolism , Adult , Female , Humans , Male , Oxygen Consumption/physiology , Physical Endurance/physiology , Sex Characteristics , Transcription Factors , Young Adult
9.
Curr Opin Genet Dev ; 38: 127-132, 2016 06.
Article in English | MEDLINE | ID: mdl-27497229

ABSTRACT

The mtDNA mutator mouse lacks the proofreading capacity of the sole mtDNA polymerase, leading to accumulation of somatic mtDNA mutations, and a profound premature aging phenotype including elevated oxidative stress and apoptosis, and reduced mitochondrial function. We have previously reported that endurance exercise alleviates the aging phenotype in the mutator mice, reduces oxidative stress, and enhances mitochondrial biogenesis. Here we summarize our findings, with the emphasis on the central role of p53 in these adaptations. We demonstrate that mtDNA in sedentary and exercised PolG mice carry similar amounts of mutations in muscle, but in addition to that sedentary mice have more non-mutational damage, which is mitigated by exercise. It follows therefore that the profound alleviation of the mtDNA mutator phenotype in muscle by exercise may not require a reduction in mtDNA mutational load, but rather a decrease of mtDNA damage and/or oxidative stress. We further hypothesize that the observed 'alleviation without a reduction of mutational load' implies that the oxidative stress in PolG muscle is maintained, at least in part, by the 'malicious cycle', a hypothetical positive feedback potentially driven by the 'transcriptional mutagenesis', that is the conversion of chemically modified nucleotides into mutant RNA bases by the mitochondrial RNA polymerase.


Subject(s)
Aging, Premature/genetics , DNA, Mitochondrial/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Tumor Suppressor Protein p53/genetics , Aging, Premature/pathology , Animals , Apoptosis/genetics , DNA Damage/genetics , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Mice , Muscle, Skeletal/metabolism , Mutation , Oxidative Stress/genetics , Physical Conditioning, Animal
10.
Nat Rev Endocrinol ; 12(9): 504-17, 2016 09.
Article in English | MEDLINE | ID: mdl-27230949

ABSTRACT

Endurance exercise-mediated multisystemic adaptations are known to mitigate metabolism-related disorders such as obesity and type 2 diabetes mellitus (T2DM). However, the underlying molecular mechanisms that promote crosstalk between organs and orchestrate the pro-metabolic effects of endurance exercise remain unclear. Exercise-induced release of peptides and nucleic acids from skeletal muscle and other organs (collectively termed 'exerkines') has been implicated in mediating these systemic adaptations. Given that the extracellular milieu is probably not a hospitable environment for labile exerkines, a lipid vehicle-based mode of delivery has originated over the course of evolution. Two types of extracellular vesicles, exosomes and microvesicles, have been shown to contain proteins and nucleic acids that participate in a variety of physiological and pathological processes. Exosomes, in particular, have been shown to facilitate the exchange of peptides, microRNA, mRNA and mitochondrial DNA between cells and tissues. Intriguingly, circulatory extracellular vesicle content increases in an intensity-dependant manner in response to endurance exercise. We propose that the systemic benefits of exercise are modulated by exosomes and/or microvesicles functioning in an autocrine, paracrine and/or endocrine manner. Furthermore, we posit that native or modified exosomes, and/or microvesicles enriched with exerkines will have therapeutic utility in the treatment of obesity and T2DM.


Subject(s)
Exercise/physiology , Exosomes/metabolism , Exosomes/transplantation , Metabolic Diseases/metabolism , Metabolic Diseases/therapy , Physical Endurance/physiology , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Muscle, Skeletal/metabolism , Obesity/metabolism , Obesity/therapy
11.
Skelet Muscle ; 6: 7, 2016.
Article in English | MEDLINE | ID: mdl-26834962

ABSTRACT

BACKGROUND: Human genetic disorders and transgenic mouse models have shown that mitochondrial DNA (mtDNA) mutations and telomere dysfunction instigate the aging process. Epidemiologically, exercise is associated with greater life expectancy and reduced risk of chronic diseases. While the beneficial effects of exercise are well established, the molecular mechanisms instigating these observations remain unclear. RESULTS: Endurance exercise reduces mtDNA mutation burden, alleviates multisystem pathology, and increases lifespan of the mutator mice, with proofreading deficient mitochondrial polymerase gamma (POLG1). We report evidence for a POLG1-independent mtDNA repair pathway mediated by exercise, a surprising notion as POLG1 is canonically considered to be the sole mtDNA repair enzyme. Here, we show that the tumor suppressor protein p53 translocates to mitochondria and facilitates mtDNA mutation repair and mitochondrial biogenesis in response to endurance exercise. Indeed, in mutator mice with muscle-specific deletion of p53, exercise failed to prevent mtDNA mutations, induce mitochondrial biogenesis, preserve mitochondrial morphology, reverse sarcopenia, or mitigate premature mortality. CONCLUSIONS: Our data establish a new role for p53 in exercise-mediated maintenance of the mtDNA genome and present mitochondrially targeted p53 as a novel therapeutic modality for diseases of mitochondrial etiology.


Subject(s)
DNA Repair , DNA, Mitochondrial/genetics , Mitochondria, Heart/metabolism , Mitochondria, Muscle/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Mutation , Myocardium/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cells, Cultured , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/genetics , Genotype , Life Expectancy , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mitochondria, Heart/pathology , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Myocardial Contraction , Myocardium/pathology , Organelle Biogenesis , Oxidative Stress , Phenotype , Protein Transport , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , Time Factors , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
13.
Neurobiol Aging ; 36(11): 2972-2983, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26294258

ABSTRACT

Mitochondrial DNA (mtDNA) mutator mice express a mutated form of mtDNA polymerase gamma that results an accelerated accumulation of somatic mtDNA mutations in association with a premature aging phenotype. An exploratory metabolomic analysis of cortical metabolites in sedentary and exercised mtDNA mutator mice and wild-type littermate controls at 9-10 months of age was performed. Pathway analysis revealed deficits in the neurotransmitters acetylcholine, glutamate, and aspartate that were ameliorated by exercise. Nicotinamide adenine dinucleotide (NAD) depletion and evidence of increased poly(adenosine diphosphate-ribose) polymerase 1 (PARP1)activity were apparent in sedentary mtDNA mutator mouse cortex, along with deficits in carnitine metabolites and an upregulated antioxidant response that largely normalized with exercise. These data highlight specific pathways that are altered in the brain in association with an accelerated age-related accumulation of somatic mtDNA mutations. These results may have relevance to age-related neurodegenerative diseases associated with mitochondrial dysfunction, such as Alzheimer's disease and Parkinson's disease and provide insights into potential mechanisms of beneficial effects of exercise on brain function.


Subject(s)
Aging/genetics , Aging/metabolism , Brain/metabolism , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Mutation , Neurotransmitter Agents/metabolism , Acetylcholine/metabolism , Animals , Antioxidants/metabolism , Aspartic Acid/metabolism , Carnitine/metabolism , DNA Damage , DNA Polymerase gamma , Female , Glutamates/metabolism , Male , Metabolomics , Mice , NAD/metabolism , Physical Conditioning, Animal , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism
14.
Am J Physiol Regul Integr Comp Physiol ; 308(8): R734-41, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25695287

ABSTRACT

Mitochondrial dysfunction may contribute to age-associated muscle atrophy. Previous data has shown that resistance exercise (RE) increases mitochondrial gene expression and enzyme activity in older adults; however, the acute response to RE has not been well characterized. To characterize the acute mitochondrial response to unaccustomed RE, healthy young (21 ± 3 yr) and older (70 ± 4 yr) men performed a unilateral RE bout for the knee extensors. Muscle biopsies were taken at rest and 3, 24, and 48 h following leg press and knee extension exercise. The expression of the mitochondrial transcriptional regulator proliferator-activated receptor γ coactivator 1-α (PGC-1α) mRNA was increased at 3 h postexercise; however, all other mitochondrial variables decreased over the postexercise period, irrespective of age. ND1, ND4, and citrate synthase (CS) mRNA were all lower at 48 h postexercise, along with specific protein subunits of complex II, III, IV, and ATP synthase. Mitochondrial DNA (mtDNA) copy number decreased by 48 h postexercise, and mtDNA deletions were higher in the older adults and remained unaffected by acute exercise. Elevated mitophagy could not explain the reduction in mitochondrial proteins and DNA, because there was no increase in ubiquitinated voltage-dependent anion channel (VDAC) or its association with PTEN-induced putative kinase 1 (Pink1) or Parkin, and elevated p62 content indicated an impairment or reduction in autophagocytic flux. In conclusion, age did not influence the response of specific mitochondrial transcripts, proteins, and DNA to a bout of RE.


Subject(s)
Aging/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Resistance Training , Adolescent , Age Factors , Aged , Aging/genetics , Aging/pathology , Autophagy , Biopsy , DNA, Mitochondrial/metabolism , Gene Expression Regulation , Humans , Male , Mitochondria, Muscle/pathology , Mitochondrial Proteins/genetics , Muscle, Skeletal/pathology , Time Factors , Transcription, Genetic , Young Adult
15.
Mitochondrion ; 21: 19-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25575636

ABSTRACT

Several studies have illustrated that the polymerase gamma mutator (PolG) mice have reduced mitochondrial content secondary to systemic mitochondrial dysfunction, and subsequently a lower capacity to perform aerobic respiration and endurance exercise. We sought to delineate the extent of glycolysis as a means of energy production in the PolG mice in the absence of optimal mitochondrial function. PolG mice display an enhanced reliance on glycolysis as compared to their wild-type counterparts. This is evident by the resting hypoglycemia, higher PFK content, and elevated plasma lactate levels in the PolG mice. In vitro experiments provide further proof that PolG derived dermal fibroblasts have a higher rate of, and capacity for, glycolysis. PolG mice also have enhanced capacity to perform hepatic gluconeogenesis that is likely enhancing the Cori cycle capacity.


Subject(s)
DNA-Directed DNA Polymerase/deficiency , Energy Metabolism , Glycolysis , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Animals , DNA Polymerase gamma , Metabolic Flux Analysis , Mice, Inbred C57BL
16.
PLoS One ; 9(10): e109189, 2014.
Article in English | MEDLINE | ID: mdl-25289940

ABSTRACT

Concurrent exercise combines different modes of exercise (e.g., aerobic and resistance) into one training protocol, providing stimuli meant to increase muscle strength, aerobic capacity and mass. As disuse is associated with decrements in strength, aerobic capacity and muscle size concurrent training is an attractive modality for rehabilitation. However, interference between the signaling pathways may result in preferential improvements for one of the exercise modes. We recruited 18 young adults (10 ♂, 8 ♀) to determine if order of exercise mode during concurrent training would differentially affect gene expression, protein content and measures of strength and aerobic capacity after 2 weeks of knee-brace induced disuse. Concurrent exercise sessions were performed 3x/week for 6 weeks at gradually increasing intensities either with endurance exercise preceding (END>RES) or following (RES>END) resistance exercise. Biopsies were collected from the vastus lateralis before, 3 h after the first exercise bout and 48 h after the end of training. Concurrent exercise altered the expression of genes involved in mitochondrial biogenesis (PGC-1α, PRC, PPARγ), hypertrophy (PGC-1α4, REDD2, Rheb) and atrophy (MuRF-1, Runx1), increased electron transport chain complex protein content, citrate synthase and mitochondrial cytochrome c oxidase enzyme activity, muscle mass, maximum isometric strength and VO 2peak. However, the order in which exercise was completed (END>RES or RES>END) only affected the protein content of mitochondrial complex II subunit. In conclusion, concurrent exercise training is an effective modality for the rehabilitation of the loss of skeletal muscle mass, maximum strength, and peak aerobic capacity resulting from disuse, regardless of the order in which the modes of exercise are performed.


Subject(s)
Exercise , Gene Expression , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Female , Gene Expression Regulation , Humans , Male , Mitochondria, Muscle/enzymology , Mitochondrial Turnover , Muscle, Skeletal/anatomy & histology , Musculoskeletal Physiological Phenomena , Organ Size , Signal Transduction , Time Factors , Young Adult
17.
Free Radic Biol Med ; 75: 241-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106705

ABSTRACT

A causal role for mitochondrial dysfunction in mammalian aging is supported by recent studies of the mtDNA mutator mouse ("PolG" mouse), which harbors a defect in the proofreading-exonuclease activity of mitochondrial DNA polymerase gamma. These mice exhibit accelerated aging phenotypes characteristic of human aging, including systemic mitochondrial dysfunction, exercise intolerance, alopecia and graying of hair, curvature of the spine, and premature mortality. While mitochondrial dysfunction has been shown to cause increased oxidative stress in many systems, several groups have suggested that PolG mutator mice show no markers of oxidative damage. These mice have been presented as proof that mitochondrial dysfunction is sufficient to accelerate aging without oxidative stress. In this study, by normalizing to mitochondrial content in enriched fractions we detected increased oxidative modification of protein and DNA in PolG skeletal muscle mitochondria. We separately developed novel methods that allow simultaneous direct measurement of mtDNA replication defects and oxidative damage. Using this approach, we find evidence that suggests PolG muscle mtDNA is indeed oxidatively damaged. We also observed a significant decrease in antioxidants and expression of mitochondrial biogenesis pathway components and DNA repair enzymes in these mice, indicating an association of maladaptive gene expression with the phenotypes observed in PolG mice. Together, these findings demonstrate the presence of oxidative damage associated with the premature aging-like phenotypes induced by mitochondrial dysfunction.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Oxidative Stress/genetics , Aging/genetics , Aging, Premature/genetics , Animals , Antioxidants/metabolism , Cell Line , DNA Breaks , DNA Polymerase gamma , DNA Replication/genetics , DNA, Mitochondrial/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/genetics , Muscle, Skeletal/metabolism , Mutation , Oxidation-Reduction
18.
Aging Cell ; 13(4): 579-82, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24894296

ABSTRACT

Somatic mtDNA mutations and deletions in particular are known to clonally expand within cells, eventually reaching detrimental intracellular concentrations. The possibility that clonal expansion is a slow process taking a lifetime had prompted an idea that founder mutations of mutant clones that cause mitochondrial dysfunction in the aged tissue might have originated early in life. If, conversely, expansion was fast, founder mutations should predominantly originate later in life. This distinction is important: indeed, from which mutations should we protect ourselves - those of early development/childhood or those happening at old age? Recently, high-resolution data describing the distribution of mtDNA deletions have been obtained using a novel, highly efficient method (Taylor et al., ). These data have been interpreted as supporting predominantly early origin of founder mutations. Re-analysis of the data implies that the data actually better fit mostly late origin of founders, although more research is clearly needed to resolve the controversy.


Subject(s)
Brain/metabolism , DNA Mutational Analysis/methods , DNA, Mitochondrial/genetics , Sequence Deletion , Humans
20.
J Biol Chem ; 289(13): 8810-7, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24505137

ABSTRACT

The transcriptional coactivator peroxisome proliferator-activator receptor γ coactivator (PGC)-1α is required for full hypoxic induction of vascular endothelial growth factor (VEGF) in skeletal muscle cells. Under normoxic conditions, PGC-1α also strongly induces mitochondrial biogenesis, but PGC-1α does not activate this program under hypoxic conditions. How this specificity is achieved is not known. We show here that hypoxia specifically induces alternatively spliced species encoding for truncated forms of PGC-1α: NT-PGC-1α and PGC-1α4. NT-PGC-1α strongly induces VEGF expression, whereas having little effect on mitochondrial genes. Conditioned medium from cells expressing NT-PGC-1α robustly induces endothelial migration and tube formation, hallmarks of angiogenesis. Transgenic expression of PGC-1α4 in skeletal muscle in mice induces angiogenesis in vivo. Finally, knockdown of these PGC-1α isoforms and hypoxia-inducible factor-1α (HIF-1α) abrogates the induction of VEGF in response to hypoxia. NT-PGC-1α and/or PGC-1α4 thus confer angiogenic specificity to the PGC-1α-mediated hypoxic response in skeletal muscle cells.


Subject(s)
Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Neovascularization, Physiologic , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Alternative Splicing , Animals , Cell Hypoxia , Cell Line , Exons/genetics , Humans , Mice , Mitochondria/genetics , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...