Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Aging Cell ; 22(6): e13827, 2023 06.
Article in English | MEDLINE | ID: mdl-37060190

ABSTRACT

Obesity significantly decreases life expectancy and increases the incidence of age-related dysfunctions, including ß-cell dysregulation leading to inadequate insulin secretion. Here, we show that diluted plasma from obese human donors acutely impairs ß-cell integrity and insulin secretion relative to plasma from lean subjects. Similar results were observed with diluted sera from obese rats fed ad libitum, when compared to sera from lean, calorically restricted, animals. The damaging effects of obese circulating factors on ß-cells occurs in the absence of nutrient overload, and mechanistically involves mitochondrial dysfunction, limiting glucose-supported oxidative phosphorylation and ATP production. We demonstrate that increased levels of adiponectin, as found in lean plasma, are the protective characteristic preserving ß-cell function; indeed, sera from adiponectin knockout mice limits ß-cell metabolic fluxes relative to controls. Furthermore, oxidative phosphorylation and glucose-sensitive insulin secretion, which are completely abrogated in the absence of this hormone, are restored by the presence of adiponectin alone, surprisingly even in the absence of other serological components, for both the insulin-secreting INS1 cell line and primary islets. The addition of adiponectin to cells treated with plasma from obese donors completely restored ß-cell functional integrity, indicating the lack of this hormone was causative of the dysfunction. Overall, our results demonstrate that low circulating adiponectin is a key damaging element for ß-cells, and suggest strong therapeutic potential for the modulation of the adiponectin signaling pathway in the prevention of age-related ß-cell dysfunction.


Subject(s)
Insulin Resistance , Insulin-Secreting Cells , Mice , Humans , Rats , Animals , Adiponectin/metabolism , Insulin Secretion , Insulin/metabolism , Obesity/metabolism , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Insulin Resistance/physiology
2.
J Biol Chem ; 299(3): 102904, 2023 03.
Article in English | MEDLINE | ID: mdl-36642177

ABSTRACT

Calcium (Ca2+) is a key regulator in diverse intracellular signaling pathways and has long been implicated in metabolic control and mitochondrial function. Mitochondria can actively take up large amounts of Ca2+, thereby acting as important intracellular Ca2+ buffers and affecting cytosolic Ca2+ transients. Excessive mitochondrial matrix Ca2+ is known to be deleterious due to opening of the mitochondrial permeability transition pore (mPTP) and consequent membrane potential dissipation, leading to mitochondrial swelling, rupture, and cell death. Moderate Ca2+ within the organelle, on the other hand, can directly or indirectly activate mitochondrial matrix enzymes, possibly impacting on ATP production. Here, we aimed to determine in a quantitative manner if extra- or intramitochondrial Ca2+ modulates oxidative phosphorylation in mouse liver mitochondria and intact hepatocyte cell lines. To do so, we monitored the effects of more modest versus supraphysiological increases in cytosolic and mitochondrial Ca2+ on oxygen consumption rates. Isolated mitochondria present increased respiratory control ratios (a measure of oxidative phosphorylation efficiency) when incubated with low (2.4 ± 0.6 µM) and medium (22.0 ± 2.4 µM) Ca2+ concentrations in the presence of complex I-linked substrates pyruvate plus malate and α-ketoglutarate, respectively, but not complex II-linked succinate. In intact cells, both low and high cytosolic Ca2+ led to decreased respiratory rates, while ideal rates were present under physiological conditions. High Ca2+ decreased mitochondrial respiration in a substrate-dependent manner, mediated by mPTP. Overall, our results uncover a Goldilocks effect of Ca2+ on liver mitochondria, with specific "just right" concentrations that activate oxidative phosphorylation.


Subject(s)
Calcium , Mitochondria , Oxidative Phosphorylation , Animals , Mice , Calcium/metabolism , Mitochondria/metabolism
3.
Am J Physiol Renal Physiol ; 323(1): F92-F106, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35499238

ABSTRACT

Caloric restriction (CR) prevents obesity and increases resilience against pathological stimuli in laboratory rodents. At the mitochondrial level, protection promoted by CR in the brain and liver is related to higher Ca2+ uptake rates and capacities, avoiding Ca2+-induced mitochondrial permeability transition. Dietary restriction has also been shown to increase kidney resistance against damaging stimuli; if these effects are related to similar mitochondrial adaptations has not been uncovered. Here, we characterized changes in mitochondrial function in response to 6 mo of CR in rats and measured bioenergetic parameters, redox balance, and Ca2+ homeostasis. CR promoted an increase in succinate-supported mitochondrial oxygen consumption rates. Although CR prevents mitochondrial reactive oxygen species production in many tissues, in kidney, we found that mitochondrial H2O2 release was enhanced in a succinate-dependent manner. Surprisingly, and opposite to the effects observed in the brain and liver, mitochondria from CR animals were more prone to Ca2+-induced mitochondrial permeability transition, in a manner reversed by the antioxidant dithiothreitol. CR mitochondria also displayed higher Ca2+ uptake rates, which were not accompanied by changes in Ca2+ efflux rates or related to altered inner mitochondrial membrane potentials or amounts of the mitochondrial Ca2+ uniporter. Instead, increased mitochondrial Ca2+ uptake rates in CR kidneys correlated with loss of mitochondrial Ca2+ uptake protein 2 (MICU2), a mitochondrial Ca2+ uniporter modulator. Interestingly, MICU2 is also modulated by CR in the liver, suggesting that it has a broader diet-sensitive regulatory role controlling mitochondrial Ca2+ homeostasis. Together, our results highlight the organ-specific bioenergetic, redox, and ionic transport results of CR, with some unexpected deleterious effects in the kidney.NEW & NOTEWORTHY Prevention of obesity through caloric restriction (CR) is well known to protect many tissues but has been poorly studied in kidneys. Here, we determined the effects of long-term CR in rat kidney mitochondria, which are central players in energy metabolism and aging. Surprisingly, we found that the diet increased mitochondrial reactive oxygen production and permeability transition. This suggests that the kidneys respond differently to restricted diets and may be more susceptible under CR.


Subject(s)
Caloric Restriction , Hydrogen Peroxide , Animals , Hydrogen Peroxide/metabolism , Kidney/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Rats , Succinates/metabolism
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(6): 166371, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35218894

ABSTRACT

Cardiovascular manifestations account for marked morbi-mortality in autosomal dominant polycystic kidney disease (ADPKD). Pkd1- and Pkd2-deficient mice develop cardiac dysfunction, however the underlying mechanisms remain largely unclear. It is unknown whether impairment of polycystin-1 cleavage at the G-protein-coupled receptor proteolysis site, a significant ADPKD mutational mechanism, is involved in this process. We analyzed the impact of polycystin-1 cleavage on heart metabolism using Pkd1V/V mice, a model unable to cleave this protein and with early cardiac dysfunction. Pkd1V/V hearts showed lower levels of glucose and amino acids and higher lipid levels than wild-types, as well as downregulation of p-AMPK, p-ACCß, CPT1B-Cpt1b, Ppara, Nppa and Acta1. These findings suggested decreased fatty acid ß-oxidation, which was confirmed by lower oxygen consumption by Pkd1V/V isolated mitochondria using palmitoyl-CoA. Pkd1V/V hearts also presented increased oxygen consumption in response to glucose, suggesting that alternative substrates may be used to generate energy. Pkd1V/V hearts displayed a higher density of decreased-size mitochondria, a finding associated with lower MFN1, Parkin and BNIP3 expression. These derangements were correlated with increased apoptosis and inflammation but not hypertrophy. Notably, Pkd1V/V neonate cardiomyocytes also displayed shifts in oxygen consumption and p-AMPK downregulation, suggesting that, at least partially, the metabolic alterations are not induced by kidney dysfunction. Our findings reveal that disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice, expanding the understanding of heart dysfunction associated with Pkd1 deficiency and likely with human ADPKD.


Subject(s)
Polycystic Kidney, Autosomal Dominant , TRPP Cation Channels , Animals , Heart , Mice , Mitochondria/metabolism , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
5.
J Bioenerg Biomembr ; 52(4): 269-277, 2020 08.
Article in English | MEDLINE | ID: mdl-32462240

ABSTRACT

Caloric restriction (CR) is widely known to increase life span and resistance to different types of injuries in several organisms. We have previously shown that mitochondria from livers or brains of CR animals exhibit higher calcium uptake rates and lower sensitivity to calcium-induced mitochondrial permeability transition (mPT), an event related to the resilient phenotype exhibited by these organs. Given the importance of calcium in metabolic control and cell homeostasis, we aimed here to uncover possible changes in mitochondrial calcium handling, redox balance and bioenergetics in cardiac and skeletal muscle mitochondria in response to six months of CR. Unexpectedly, we found that CR does not alter the susceptibility to mPT in muscle (cardiac or skeletal), nor calcium uptake rates. Despite the lack in changes in calcium transport properties, CR consistently decreased respiration in the presence of ATP synthesis in heart and soleus muscle. In heart, such changes were accompanied by a decrease in respiration in the absence of ATP synthesis, lower maximal respiratory rates and a reduced rate of hydrogen peroxide release. Hydrogen peroxide release was unaltered by CR in skeletal muscle. No changes were observed in inner membrane potentials and respiratory control ratios. Together, these results highlight the tissue-specific bioenergetic and ion transport effects induced by CR, demonstrating that resilience against calcium-induced mPT is not present in all tissues.


Subject(s)
Caloric Restriction/standards , Energy Metabolism/physiology , Muscle, Skeletal/immunology , Myocardium/immunology , Animals , Male , Rats , Rats, Sprague-Dawley
6.
Biosci Rep ; 39(4)2019 04 30.
Article in English | MEDLINE | ID: mdl-30867255

ABSTRACT

Microglial activation by oleate and palmitate differentially modulates brain inflammatory status. However, the metabolic reprogramming supporting these reactive phenotypes remains unknown. Employing real-time metabolic measurements and lipidomic analysis, we show that both fatty acids promote microglial oxidative metabolism, while lipopolysaccharide (LPS) enhances glycolytic rates. Interestingly, oleate treatment was followed by enrichment in storage lipids bound to polyunsaturated fatty acids (PUFA), in parallel with protection against oxidative imbalance. Palmitate, in turn, induced a distinct lipid distribution defined by PUFA linked to membrane phospholipids, which are more susceptible to lipid peroxidation and inflammatory signaling cascades. This distribution was mirrored by LPS treatment, which led to a strong pro-inflammatory phenotype in microglia. Thus, although both oleate and palmitate preserve mitochondrial function, a contrasting lipid distribution supports differences in fatty acid-induced neuroinflammation. These data reinforce the concept that reactive microglial profiles are achieved by stimulus-evoked remodeling in cell metabolism.


Subject(s)
Brain/pathology , Microglia/metabolism , Mitochondria/metabolism , Oleic Acid/metabolism , Palmitates/metabolism , Animals , Cell Line , Glycolysis/physiology , Inflammation/pathology , Lipid Peroxidation/physiology , Lipopolysaccharides/pharmacology , Membrane Lipids/metabolism , Mice , Microglia/cytology , Neuroimmunomodulation/physiology , Oxidation-Reduction , Oxidative Phosphorylation
7.
PLoS One ; 14(2): e0211733, 2019.
Article in English | MEDLINE | ID: mdl-30716103

ABSTRACT

Obesity-derived inflammation and metabolic dysfunction has been related to the activity of the inducible nitric oxide synthase (iNOS). To understand the interrelation between metabolism, obesity and NO., we evaluated the effects of obesity-induced NO. signaling on liver mitochondrial function. We used mouse strains containing mitochondrial nicotinamide transhydrogenase activity, while prior studies involved a spontaneous mutant of this enzyme, and are, therefore, more prone to oxidative imbalance. Wild-type and iNOS knockout mice were fed a high fat diet for 2, 4 or 8 weeks. iNOS knockout did not protect against diet-induced metabolic changes. However, the diet decreased fatty-acid oxidation capacity in liver mitochondria at 4 weeks in both wild-type and knockout groups; this was recovered at 8 weeks. Interestingly, other mitochondrial functional parameters were unchanged, despite significant modifications in insulin resistance in wild type and iNOS knockout animals. Overall, we found two surprising features of obesity-induced metabolic dysfunction: (i) iNOS does not have an essential role in obesity-induced insulin resistance under all experimental conditions and (ii) liver mitochondria are resilient to functional changes in obesity-induced metabolic dysfunction.


Subject(s)
Insulin Resistance/physiology , Liver/metabolism , Liver/physiology , Mitochondria, Liver/metabolism , Mitochondria, Liver/physiology , Nitric Oxide Synthase Type II/metabolism , Animals , Diet, High-Fat , Lipid Metabolism/physiology , Male , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/physiopathology
8.
Fungal Biol ; 123(1): 59-65, 2019 01.
Article in English | MEDLINE | ID: mdl-30654958

ABSTRACT

Blastocladiella emersonii is an early diverging fungus of the phylum Blastocladiomycota. During the life cycle of the fungus, mitochondrial morphology changes significantly, from a fragmented form in sessile vegetative cells to a fused network in motile zoospores. In this study, we visualize these morphological changes using a mitochondrial fluorescent probe and show that the respiratory capacity in zoospores is much higher than in vegetative cells, suggesting that mitochondrial morphology could be related to the differences in oxygen consumption. While studying the respiratory chain of the fungus, we observed an antimycin A and cyanide-insensitive, salicylhydroxamic (SHAM)-sensitive respiratory activity, indicative of a mitochondrial alternative oxidase (AOX) activity. The presence of AOX was confirmed by the finding of a B. emersonii cDNA encoding a putative AOX, and by detection of AOX protein in immunoblots. Inhibition of AOX activity by SHAM was found to significantly alter the capacity of the fungus to grow and sporulate, indicating that AOX participates in life cycle control in B. emersonii.


Subject(s)
Blastocladiella/growth & development , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Spores, Fungal/growth & development , Electron Transport , Oxidation-Reduction , Oxygen/metabolism
9.
Endocrinology ; 155(7): 2456-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24797627

ABSTRACT

Intermittent fasting (IF) is an often-used intervention to decrease body mass. In male Sprague-Dawley rats, 24 hour cycles of IF result in light caloric restriction, reduced body mass gain, and significant decreases in the efficiency of energy conversion. Here, we study the metabolic effects of IF in order to uncover mechanisms involved in this lower energy conversion efficiency. After 3 weeks, IF animals displayed overeating during fed periods and lower body mass, accompanied by alterations in energy-related tissue mass. The lower efficiency of energy use was not due to uncoupling of muscle mitochondria. Enhanced lipid oxidation was observed during fasting days, whereas fed days were accompanied by higher metabolic rates. Furthermore, an increased expression of orexigenic neurotransmitters AGRP and NPY in the hypothalamus of IF animals was found, even on feeding days, which could explain the overeating pattern. Together, these effects provide a mechanistic explanation for the lower efficiency of energy conversion observed. Overall, we find that IF promotes changes in hypothalamic function that explain differences in body mass and caloric intake.


Subject(s)
Body Weight/physiology , Fasting/physiology , Feeding Behavior/physiology , Hyperphagia/physiopathology , Hypothalamus/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Eating/drug effects , Energy Metabolism/physiology , Gene Expression , Immunoblotting , Leptin/pharmacology , Lipid Metabolism/physiology , Male , Mitochondria, Muscle/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Time Factors
10.
J Bioenerg Biomembr ; 43(5): 531-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21833602

ABSTRACT

Energy metabolism in the adult brain consumes large quantities of glucose, but little is known to date regarding how glucose metabolism changes during neuronal differentiation, a process that is highly demanding energetically. We studied changes in glucose metabolism during neuronal differentiation of P19 mouse embryonal carcinoma cells, E14Tg2A embryonic stem cells as well as during brain development of BLC57 mice. In all these models, we find that neurogenesis is accompanied by a shift from oxidative to fermentative glucose metabolism. This shift is accompanied by both a decrease in mitochondrial enzymatic activities and mitochondrial uncoupling. In keeping with this finding, we also observe that differentiation does not require oxidative metabolism, as indicated by experiments demonstrating that the process is preserved in cells treated with the ATP synthase inhibitor oligomycin. Overall, we provide evidence that neuronal differentiation involves a shift from oxidative to fermentative metabolism, and that oxidative phosphorylation is not essential for this process.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Glucose/metabolism , Neurons/metabolism , Animals , Brain/embryology , Brain/metabolism , Cell Line, Tumor , Embryonic Stem Cells/cytology , Enzyme Inhibitors/pharmacology , Mice , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/metabolism , Neurons/cytology , Oligomycins/pharmacology , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects
11.
Free Radic Biol Med ; 51(7): 1454-60, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21816219

ABSTRACT

Calorie restriction is a dietary intervention known to improve redox state, glucose tolerance, and animal life span. Other interventions have been adopted as study models for caloric restriction, including nonsupplemented food restriction and intermittent, every-other-day feedings. We compared the short- and long-term effects of these interventions to ad libitum protocols and found that, although all restricted diets decrease body weight, intermittent feeding did not decrease intra-abdominal adiposity. Short-term calorie restriction and intermittent feeding presented similar results relative to glucose tolerance. Surprisingly, long-term intermittent feeding promoted glucose intolerance, without a loss in insulin receptor phosphorylation. Intermittent feeding substantially increased insulin receptor nitration in both intra-abdominal adipose tissue and muscle, a modification associated with receptor inactivation. All restricted diets enhanced nitric oxide synthase levels in the insulin-responsive adipose tissue and skeletal muscle. However, whereas calorie restriction improved tissue redox state, food restriction and intermittent feedings did not. In fact, long-term intermittent feeding resulted in largely enhanced tissue release of oxidants. Overall, our results show that restricted diets are significantly different in their effects on glucose tolerance and redox state when adopted long-term. Furthermore, we show that intermittent feeding can lead to oxidative insulin receptor inactivation and glucose intolerance.


Subject(s)
Caloric Restriction/methods , Diet, Reducing/methods , Intra-Abdominal Fat/metabolism , Muscle, Skeletal/metabolism , Obesity/diet therapy , Receptor, Insulin/metabolism , Adiposity , Animals , Blotting, Western , Body Weight , Glucose/metabolism , Glucose Intolerance/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/analysis , Insulin Receptor Substrate Proteins/biosynthesis , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/biosynthesis , Nitro Compounds , Obesity/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Receptor, Insulin/antagonists & inhibitors
12.
Curr Drug Targets ; 12(6): 783-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21275885

ABSTRACT

Mild mitochondrial uncoupling, or the reduction of the efficiency of energy conversion without compromising intracellular high energy phosphate levels, is a protective therapeutic strategy under many laboratory conditions. Here we discuss these conditions, which include both cell and animal models of ischemia reperfusion and complications associated with the metabolic syndrome. We also discuss drugs that promote mild mitochondrial uncoupling and naturally occurring mild mitochondrial uncoupling pathways involving free fatty acid cycling and K(+) transport.


Subject(s)
Metabolic Syndrome/complications , Mitochondria/drug effects , Reperfusion Injury/physiopathology , Animals , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Humans , Ion Channels/metabolism , Ion Transport , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Potassium/metabolism , Uncoupling Protein 1
13.
PLoS One ; 5(5): e10666, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20498724

ABSTRACT

BACKGROUND: The aim of the present study was to evaluate the protective effects of the 4-anilinoquinazoline derivative PD153035 on cardiac ischemia/reperfusion and mitochondrial function. METHODOLOGY/PRINCIPAL FINDINGS: Perfused rat hearts and cardiac HL-1 cells were used to determine cardioprotective effects of PD153035. Isolated rat heart mitochondria were studied to uncover mechanisms of cardioprotection. Nanomolar doses of PD153035 strongly protect against heart and cardiomyocyte damage induced by ischemia/reperfusion and cyanide/aglycemia. PD153035 did not alter oxidative phosphorylation, nor directly prevent Ca(2+) induced mitochondrial membrane permeability transition. The protective effect of PD153035 on HL-1 cells was also independent of AKT phosphorylation state. Interestingly, PD153035 activated K(+) transport in isolated mitochondria, in a manner prevented by ATP and 5-hydroxydecanoate, inhibitors of mitochondrial ATP-sensitive K(+) channels (mitoK(ATP)). 5-Hydroxydecanoate also inhibited the cardioprotective effect of PD153035 in cardiac HL-1 cells, demonstrating that this protection is dependent on mitoK(ATP) activation. CONCLUSIONS/SIGNIFICANCE: We conclude that PD153035 is a potent cardioprotective compound and acts in a mechanism involving mitoK(ATP) activation.


Subject(s)
Cardiotonic Agents/pharmacology , Ion Channel Gating/drug effects , Potassium Channels/metabolism , Quinazolines/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Biological Transport/drug effects , Calcium/metabolism , Cell Respiration/drug effects , Cyanides/toxicity , Heart Function Tests , In Vitro Techniques , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Permeability/drug effects , Phosphorylation/drug effects , Potassium/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
14.
Aging Cell ; 7(4): 552-60, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18505478

ABSTRACT

Caloric restriction is the most effective non-genetic intervention to enhance lifespan known to date. A major research interest has been the development of therapeutic strategies capable of promoting the beneficial results of this dietary regimen. In this sense, we propose that compounds that decrease the efficiency of energy conversion, such as mitochondrial uncouplers, can be caloric restriction mimetics. Treatment of mice with low doses of the protonophore 2,4-dinitrophenol promotes enhanced tissue respiratory rates, improved serological glucose, triglyceride and insulin levels, decrease of reactive oxygen species levels and tissue DNA and protein oxidation, as well as reduced body weight. Importantly, 2,4-dinitrophenol-treated animals also presented enhanced longevity. Our results demonstrate that mild mitochondrial uncoupling is a highly effective in vivo antioxidant strategy, and describe the first therapeutic intervention capable of effectively reproducing the physiological, metabolic and lifespan effects of caloric restriction in healthy mammals.


Subject(s)
2,4-Dinitrophenol/pharmacology , Energy Metabolism/drug effects , Longevity/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Uncoupling Agents/pharmacology , Animals , Blood Glucose/metabolism , Cell Respiration/drug effects , Female , Insulin/blood , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Triglycerides/blood
15.
Oncogene ; 23(39): 6684-92, 2004 Aug 26.
Article in English | MEDLINE | ID: mdl-15221013

ABSTRACT

A large fraction of transcripts are expressed antisense to introns of known genes in the human genome. Here we show the construction and use of a cDNA microarray platform enriched in intronic transcripts to assess their biological relevance in pathological conditions. To validate the approach, prostate cancer was used as a model, and 27 patient tumor samples with Gleason scores ranging from 5 to 10 were analyzed. We find that a considerably higher fraction (6.6%, [23/346]) of intronic transcripts are significantly correlated (P< or =0.001) to the degree of prostate tumor differentiation (Gleason score) when compared to transcripts from unannotated genomic regions (1%, [6/539]) or from exons of known genes (2%, [27/1369]). Among the top twelve transcripts most correlated to tumor differentiation, six are antisense intronic messages as shown by orientation-specific RT-PCR or Northern blot analysis with strand-specific riboprobe. Orientation-specific real-time RT-PCR with six tumor samples, confirmed the correlation (P=0.024) between the low/high degrees of tumor differentiation and antisense intronic RASSF1 transcript levels. The need to use intron arrays to reveal the transcriptome profile of antisense intronic RNA in cancer has clearly emerged.


Subject(s)
Cell Differentiation/genetics , Introns , Prostatic Neoplasms/pathology , RNA, Antisense/metabolism , Humans , Male , Molecular Sequence Data , Polymerase Chain Reaction , Prostatic Neoplasms/genetics , RNA, Antisense/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...