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.
Diabetes ; 65(9): 2772-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27335231

ABSTRACT

Hypersecretion of amylin is common in individuals with prediabetes, causes amylin deposition and proteotoxicity in pancreatic islets, and contributes to the development of type 2 diabetes. Recent studies also identified amylin deposits in failing hearts from patients with obesity or type 2 diabetes and demonstrated that hyperamylinemia accelerates the development of heart dysfunction in rats expressing human amylin in pancreatic ß-cells (HIP rats). To further determine the impact of hyperamylinemia on cardiac myocytes, we investigated human myocardium, compared diabetic HIP rats with diabetic rats expressing endogenous (nonamyloidogenic) rat amylin, studied normal mice injected with aggregated human amylin, and developed in vitro cell models. We found that amylin deposition negatively affects cardiac myocytes by inducing sarcolemmal injury, generating reactive aldehydes, forming amylin-based adducts with reactive aldehydes, and increasing synthesis of the proinflammatory cytokine interleukin-1ß (IL-1ß) independently of hyperglycemia. These results are consistent with the pathological role of amylin deposition in the pancreas, uncover a novel contributing mechanism to cardiac myocyte injury in type 2 diabetes, and suggest a potentially treatable link of type 2 diabetes with diabetic heart disease. Although further studies are necessary, these data also suggest that IL-1ß might function as a sensor of myocyte amylin uptake and a potential mediator of myocyte injury.


Subject(s)
Interleukin-1beta/metabolism , Islet Amyloid Polypeptide/metabolism , Lipid Peroxides/metabolism , Myocardium/metabolism , Sarcolemma/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Immunochemistry , Metabolomics , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
2.
J Alzheimers Dis ; 53(1): 259-72, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27163815

ABSTRACT

Amylin is a hormone synthesized and co-secreted with insulin by pancreatic ß-cells that crosses the blood-brain barrier and regulates satiety. Amylin from humans (but not rodents) has an increased propensity to aggregate into pancreatic islet amyloid deposits that contribute to ß-cell mass depletion and development of type-2 diabetes by inducing oxidative stress and inflammation. Recent studies demonstrated that aggregated amylin also accumulates in brains of Alzheimer's disease (AD) patients, preponderantly those with type-2 diabetes. Here, we report that, in addition to amylin plaques and mixed amylin-Aß deposits, brains of diabetic patients with AD show amylin immunoreactive deposits inside the neurons. Neuronal amylin formed adducts with 4-hydroxynonenal (4-HNE), a marker of peroxidative membrane injury, and increased synthesis of the proinflammatory cytokine interleukin (IL)-1ß. These pathological changes were mirrored in rats expressing human amylin in pancreatic islets (HIP rats) and mice intravenously injected with aggregated human amylin, but not in hyperglycemic rats secreting wild-type non-amyloidogenic rat amylin. In cultured primary hippocampal rat neurons, aggregated amylin increased IL-1ß synthesis via membrane destabilization and subsequent generation of 4-HNE. These effects were blocked by membrane stabilizers and lipid peroxidation inhibitors. Thus, elevated circulating levels of aggregated amylin negatively affect the neurons causing peroxidative membrane injury and aberrant inflammatory responses independent of other confounding factors of diabetes. The present results are consistent with the pathological role of aggregated amylin in the pancreas, demonstrate a novel contributing mechanism to neurodegeneration, and suggest a direct, potentially treatable link of type-2 diabetes with AD.


Subject(s)
Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Diabetes Mellitus, Type 2/pathology , Interleukin-1beta/metabolism , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans/metabolism , Aged , Aged, 80 and over , Aldehydes/metabolism , Alzheimer Disease/complications , Animals , Animals, Newborn , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Fasting/physiology , Female , Hippocampus/cytology , Humans , Islet Amyloid Polypeptide/pharmacology , Ligation , Lipid Peroxidation/physiology , Male , Mice , Rats , Rats, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...