Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Nucl Med ; 59(3): 516-522, 2018 03.
Article in English | MEDLINE | ID: mdl-28912148

ABSTRACT

Obesity and its metabolic consequences are a major cause of morbidity and mortality. Brown adipose tissue (BAT) utilizes glucose and free fatty acids to produce heat, thereby increasing energy expenditure. Effective evaluation of human BAT stimulators is constrained by the current standard method of assessing BAT-PET/CT-as it requires exposure to high doses of ionizing radiation. Infrared thermography (IRT) is a potential noninvasive, safe alternative, although direct corroboration with PET/CT has not been established. Methods: IRT and 18F-FDG PET/CT data from 8 healthy men subjected to water-jacket cooling were directly compared. Thermal images were geometrically transformed to overlay PET/CT-derived maximum intensity projection (MIP) images from each subject, and the areas with the most intense temperature and glucose uptake within the supraclavicular regions were compared. Relationships between supraclavicular temperatures (TSCR) from IRT and the metabolic rate of glucose uptake (MR(gluc)) from PET/CT were determined. Results: Glucose uptake on MR(gluc)MIP was found to correlate positively with a change in TSCR relative to a reference region (r2 = 0.721; P = 0.008). Spatial overlap between areas of maximal MR(gluc)MIP and maximal TSCR was 29.5% ± 5.1%. Prolonged cooling, for 60 min, was associated with a further TSCR rise, compared with cooling for 10 min. Conclusion: The supraclavicular hotspot identified on IRT closely corresponded to the area of maximal uptake on PET/CT-derived MR(gluc)MIP images. Greater increases in relative TSCR were associated with raised glucose uptake. IRT should now be considered a suitable method for measuring BAT activation, especially in populations for whom PET/CT is not feasible, practical, or repeatable.


Subject(s)
Adipose Tissue, Brown/diagnostic imaging , Positron Emission Tomography Computed Tomography , Thermography , Adipose Tissue, Brown/cytology , Adolescent , Female , Humans , Male , Young Adult
2.
PLoS One ; 8(4): e61626, 2013.
Article in English | MEDLINE | ID: mdl-23620776

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive fatal neurodegenerative disease characterised by loss of motor neurons that currently has no cure. Omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid (EPA), have many health benefits including neuroprotective and myoprotective potential. We tested the hypothesis that a high level of dietary EPA could exert beneficial effects in ALS. The dietary exposure to EPA (300 mg/kg/day) in a well-established mouse model of ALS expressing the G93A superoxide dismutase 1 (SOD1) mutation was initiated at a pre-symptomatic or symptomatic stage, and the disease progression was monitored until the end stage. Daily dietary EPA exposure initiated at the disease onset did not significantly alter disease presentation and progression. In contrast, EPA treatment initiated at the pre-symptomatic stage induced a significantly shorter lifespan. In a separate group of animals sacrificed before the end stage, the tissue analysis showed that the vacuolisation detected in G93A-SOD1 mice was significantly increased by exposure to EPA. Although EPA did not alter motor neurone loss, EPA reversed the significant increase in activated microglia and the astrocytic activation seen in G93A-SOD1 mice. The microglia in the spinal cord of G93A-SOD1 mice treated with EPA showed a significant increase in 4-hydroxy-2-hexenal, a highly toxic aldehydic oxidation product of omega-3 fatty acids. These data show that dietary EPA supplementation in ALS has the potential to worsen the condition and accelerate the disease progression. This suggests that great caution should be exerted when considering dietary omega-3 fatty acid supplements in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Disease Progression , Eicosapentaenoic Acid/adverse effects , Administration, Oral , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Dietary Supplements , Disease Models, Animal , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/blood , Female , Humans , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Mutant Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Survival Analysis , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vacuoles/drug effects , Vacuoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...