Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Vet J ; 199(1): 24-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24246648

ABSTRACT

The metabolic syndrome is a set of risk factors for the development of type 2 diabetes, atherosclerosis, coronary heart disease and stroke in human beings. The term has recently been applied to dogs that exhibit components of the human metabolic syndrome, specifically visceral obesity, hypercholesterolaemia, hypertriglyceridaemia, hypertension and fasting hyperglycaemia. Obese dogs, like obese humans, are known to develop resistance to the glucose-lowering effects of insulin, and develop increased circulating concentrations of triglycerides, cholesterol and blood pressure. Unlike humans, however, obese dogs do not develop fasting hyperglycaemia or atherogenic hyperlipidaemia. Importantly, there is no evidence that dogs develop type 2 diabetes. Atherosclerosis, coronary heart disease and stroke are rare and not known to be associated with obesity in dogs. On the basis of current knowledge, the use of the term 'metabolic syndrome' in dogs does not appear to have merit.


Subject(s)
Dog Diseases/pathology , Metabolic Syndrome/veterinary , Obesity/veterinary , Animals , Dog Diseases/diagnosis , Dogs , Obesity/pathology
2.
Gen Comp Endocrinol ; 172(3): 468-74, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21530529

ABSTRACT

Adiponectin is a key adipokine that regulates carbohydrate and lipid metabolism. It circulates in stable low (LMW) and high molecular weight (HMW) forms. The aims of this study were to characterize baseline adiponectin profiles (total, LMW and HMW multimers) in healthy cats and to assess the effects of varying dietary carbohydrate content on adiponectin profiles. Cats were maintained on a diet with moderate carbohydrate content (37% metabolisable energy [ME]) for 4 weeks and then randomly allocated to either a low carbohydrate (19% ME) or high carbohydrate (52% ME) diet for 4 weeks. Fasting and postprandial plasma adiponectin profiles were measured by ELISA and sucrose gradient/Western blot. After consuming the moderate carbohydrate diet for 4 weeks, fasting total, HMW and LMW plasma adiponectin concentrations were 5.0±0.6, 2.5±0.5 and 2.6±0.2 µg/mL, respectively. After changing to the low carbohydrate diet, fasting total adiponectin was unchanged but HMW adiponectin increased and LMW adiponectin decreased. No significant postprandial changes were observed. Cats consuming the high carbohydrate diet had increased fasting total and LMW adiponectin with no change in HMW adiponectin. In the postprandial state total adiponectin was reduced and there was a trend towards a decrease in HMW (p=0.086) but not LMW multimers. These data indicate that feline adiponectin multimer profiles are similar to those reported in other species and demonstrate that changes in plasma adiponectin occur in response to chronic and acute carbohydrate intake and these reflect differential changes in adiponectin multimers.


Subject(s)
Adiponectin/blood , Cats/metabolism , Dietary Carbohydrates/metabolism , Adiponectin/chemistry , Animals , Blood Glucose/metabolism , Female , Insulin/blood , Male
3.
Am J Vet Res ; 72(3): 357-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21355739

ABSTRACT

OBJECTIVE: To compare beta-cell sensitivity to glucose, first-phase insulin secretion, and glucose tolerance between dogs with naturally occurring obesity of > 2 years' duration and lean dogs. ANIMALS: 17 client-owned obese or lean dogs. PROCEDURES: Frequently sampled IV glucose tolerance tests were performed with minimal model analysis on 6 obese dogs and matched controls. Glucagon stimulation tests were performed on 5 obese dogs and matched controls. RESULTS: Obese dogs were half as sensitive to the effects of insulin as lean dogs. Plasma glucose concentrations after food withholding did not differ significantly between groups; plasma insulin concentrations were 3 to 4 times as great in obese as in lean dogs. Obese dogs had plasma insulin concentrations twice those of lean dogs after administration of glucose and 4 times as great after administration of glucagon. First-phase insulin secretion was greater in obese dogs. CONCLUSIONS AND CLINICAL RELEVANCE: Obese dogs compensated for obesity-induced insulin resistance by secreting more insulin. First-phase insulin secretion and beta-cell glucose sensitivity were not lost despite years of obesity-induced insulin resistance and compensatory hyperinsulinemia. These findings help explain why dogs, unlike cats and humans, have not been documented to develop type 2 diabetes mellitus.


Subject(s)
Blood Glucose/metabolism , Dog Diseases/physiopathology , Glucagon/metabolism , Glucose Tolerance Test/veterinary , Insulin-Secreting Cells/physiology , Insulin/metabolism , Obesity/veterinary , Animals , Dogs , Insulin/blood , Insulin Resistance , Insulin Secretion , Obesity/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...