Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetologia ; 64(4): 865-877, 2021 04.
Article in English | MEDLINE | ID: mdl-33515072

ABSTRACT

AIMS/HYPOTHESIS: Pancreatic beta cells are subjected to exogenous damaging factors such as proinflammatory cytokines or excess glucose that can cause accumulation of damage-inducing reactive oxygen species during the pathogenesis of diabetes. We and others have shown that beta cell autophagy can reduce reactive oxygen species to protect against apoptosis. While impaired islet autophagy has been demonstrated in human type 2 diabetes, it is unknown if islet autophagy is perturbed in the pathogenesis of type 1 diabetes. We hypothesised that beta cell autophagy is dysfunctional in type 1 diabetes, and that there is a progressive loss during early diabetes development. METHODS: Pancreases were collected from chloroquine-injected and non-injected non-obese diabetes-resistant (NOR) and non-obese diabetic (NOD) mice. Age- and BMI-matched pancreas tissue sections from human organ donors (N = 34) were obtained from the Network for Pancreatic Organ Donors with Diabetes (nPOD). Tissue sections were stained with antibodies against proinsulin or insulin (beta cell markers), microtubule-associated protein 1 light chain 3 A/B (LC3A/B; autophagosome marker), lysosomal-associated membrane protein 1 (LAMP1; lysosome marker) and p62 (autophagy adaptor). Images collected on a scanning laser confocal microscope were analysed with CellProfiler and ImageJ. Secondary lysosomes and telolysosomes were assessed in electron micrographs of human pancreatic tissue sections (n = 12), and energy dispersive x-ray analysis was performed to assess distribution of elements (n = 5). RESULTS: We observed increased autophagosome numbers in islets of diabetic NOD mice (p = 0.008) and increased p62 in islets of both non-diabetic and diabetic NOD mice (p < 0.001) vs NOR mice. There was also a reduction in LC3-LAMP1 colocalisation in islets of diabetic NOD mice compared with both non-diabetic NOD (p < 0.001) and NOR mice (p < 0.001). Chloroquine elicited accumulation of autophagosomes in the islets of NOR (p = 0.003) and non-diabetic NOD mice (p < 0.001), but not in islets of diabetic NOD mice; and stimulated accumulation of p62 in NOR (p < 0.001), but not in NOD mice. We observed reduced LC3-LAMP1 colocalisation (p < 0.001) in residual beta cells of human donors with type 1 diabetes vs non-diabetic participants. We also observed reduced colocalisation of proinsulin with LAMP1 in donors with type 1 diabetes (p < 0.001). Electron microscopy also revealed accumulation of telolysosomes with nitrogen-dense rings in beta cells of autoantibody-positive donors (p = 0.002). CONCLUSIONS/INTERPRETATION: We provide evidence of islet macroautophagy/crinophagy impairment in human type 1 diabetes. We also document accumulation of telolysosomes with peripheral nitrogen in beta cells of autoantibody-positive donors, demonstrating altered lysosome content that may be associated with lysosome dysfunction before clinical hyperglycaemia. Similar macroautophagy impairments are present in the NOD mouse model of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Lysosomes/pathology , Macroautophagy , Adolescent , Adult , Animals , Autophagy-Related Proteins/metabolism , Case-Control Studies , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Female , Humans , Insulin-Secreting Cells/metabolism , Lysosomes/metabolism , Male , Mice, Inbred NOD , Signal Transduction , Young Adult
2.
Diabetes ; 67(8): 1576-1588, 2018 08.
Article in English | MEDLINE | ID: mdl-29784660

ABSTRACT

Production of reactive oxygen species (ROS) is a key instigator of ß-cell dysfunction in diabetes. The pleiotropic cytokine interleukin 6 (IL-6) has previously been linked to ß-cell autophagy but has not been studied in the context of ß-cell antioxidant response. We used a combination of animal models of diabetes and analysis of cultured human islets and rodent ß-cells to study how IL-6 influences antioxidant response. We show that IL-6 couples autophagy to antioxidant response and thereby reduces ROS in ß-cells and human islets. ß-Cell-specific loss of IL-6 signaling in vivo renders mice more susceptible to oxidative damage and cell death through the selective ß-cell toxins streptozotocin and alloxan. IL-6-driven ROS reduction is associated with an increase in the master antioxidant factor NRF2, which rapidly translocates to the mitochondria to decrease mitochondrial activity and stimulate mitophagy. IL-6 also initiates a robust transient decrease in cellular cAMP levels, likely contributing to the stimulation of mitophagy to mitigate ROS. Our findings suggest that coupling autophagy to antioxidant response in ß-cells leads to stress adaptation that can reduce cellular apoptosis. These findings have implications for ß-cell survival under diabetogenic conditions and present novel targets for therapeutic intervention.


Subject(s)
Autophagy , Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-6/metabolism , Oxidative Stress , Receptors, Interleukin-6/agonists , Signal Transduction , Alloxan/toxicity , Animals , Autophagy/drug effects , Biomarkers/metabolism , Cell Line, Tumor , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Interleukin-6/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress/drug effects , Random Allocation , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Streptozocin/toxicity , Tissue Banks , Tissue Culture Techniques
3.
Endocrinology ; 159(5): 2127-2141, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29617763

ABSTRACT

Nearly 100 years have passed since Frederick Banting and Charles Best first discovered and purified insulin. Their discovery and subsequent improvements revolutionized the treatment of diabetes, and the field continues to move at an ever-faster pace with respect to unique treatments for both type 1 and type 2 diabetes. Despite these advances, we still do not fully understand how apoptosis of the insulin-producing ß-cells is triggered, presenting a challenge in the development of preventative measures. In recent years, the process of autophagy has generated substantial interest in this realm due to discoveries highlighting its clear role in the maintenance of cellular homeostasis. As a result, the number of studies focused on islet and ß-cell autophagy has increased substantially in recent years. In this review, we will discuss what is currently known regarding the role of ß-cell autophagy in type 1 and type 2 diabetes pathogenesis, with an emphasis on new and exciting developments over the past 5 years. Further, we will discuss how these discoveries might be translated into unique treatments in the coming years.


Subject(s)
Autophagy/physiology , Diabetes Mellitus/metabolism , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/metabolism , Oxidative Stress , Animals , Apoptosis , Cell Survival , Cellular Reprogramming Techniques , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Diabetes Mellitus/therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diet Therapy , Feedback, Physiological , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Vitamin D/therapeutic use , Vitamins/therapeutic use
4.
FASEB J ; 31(9): 4140-4152, 2017 09.
Article in English | MEDLINE | ID: mdl-28592636

ABSTRACT

IL-6 is a pleiotropic cytokine with complex roles in inflammation and metabolic disease. The role of IL-6 as a pro- or anti-inflammatory cytokine is still unclear. Within the pancreatic islet, IL-6 stimulates secretion of the prosurvival incretin hormone glucagon-like peptide 1 (GLP-1) by α cells and acts directly on ß cells to stimulate insulin secretion in vitro Uncovering physiologic mechanisms promoting ß-cell survival under conditions of inflammation and stress can identify important pathways for diabetes prevention and treatment. Given the established role of GLP-1 in promoting ß-cell survival, we hypothesized that IL-6 may also directly protect ß cells from apoptosis. Herein, we show that IL-6 robustly activates signal transducer and activator of transcription 3 (STAT3), a transcription factor that is involved in autophagy. IL-6 stimulates LC3 conversion and autophagosome formation in cultured ß cells. In vivo IL-6 infusion stimulates a robust increase in lysosomes in the pancreas that is restricted to the islet. Autophagy is critical for ß-cell homeostasis, particularly under conditions of stress and increased insulin demand. The stimulation of autophagy by IL-6 is regulated via multiple complementary mechanisms including inhibition of mammalian target of rapamycin complex 1 (mTORC1) and activation of Akt, ultimately leading to increases in autophagy enzyme production. Pretreatment with IL-6 renders ß cells resistant to apoptosis induced by proinflammatory cytokines, and inhibition of autophagy with chloroquine prevents the ability of IL-6 to protect from apoptosis. Importantly, we find that IL-6 can activate STAT3 and the autophagy enzyme GABARAPL1 in human islets. We also see evidence of decreased IL-6 pathway signaling in islets from donors with type 2 diabetes. On the basis of our results, we propose direct stimulation of autophagy as a novel mechanism for IL-6-mediated protection of ß cells from stress-induced apoptosis.-Linnemann, A. K., Blumer, J., Marasco, M. R., Battiola, T. J., Umhoefer, H. M., Han, J. Y., Lamming, D. W., Davis, D. B. Interleukin 6 protects pancreatic ß cells from apoptosis by stimulation of autophagy.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Insulin-Secreting Cells/metabolism , Interleukin-6/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2 , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Infusion Pumps, Implantable , Interleukin-6/genetics , Interleukin-6/pharmacology , Islets of Langerhans/metabolism , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Rats , Recombinant Proteins , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...