5 research outputs found

    Limited impact on glucose homeostasis of leptin receptor deletion from insulin- or proglucagon-expressing cells

    Get PDF
    Aims/hypothesis The adipose tissue-derived hormone leptin plays an important role in the maintenance of body weight and glucose homeostasis. Leptin mediates its effects by interaction with leptin receptors (LepRb), which are highly expressed in the hypothalamus and other brain centres, and at lower levels in the periphery. Previous studies have used relatively promiscuous or inefficient Cre deleter strains, respectively, to explore the roles of LepR in pancreatic β and α cells. Here, we use two newly-developed Cre lines to explore the role of leptin signalling in insulin and proglucagon-expressing cells. Methods Leptin receptor expression was measured in isolated mouse islets and highly-purified islet cells by RNASeq and quantitative RT-PCR. Mice lacking leptin signalling in pancreatic β, or in α and other proglucagon-expressing cells, were generated using Ins1Cre- or iGluCre-mediated recombination respectively of flox'd leptin receptor alleles. In vivo glucose homeostasis, changes in body weight, pancreatic histology and hormone secretion from isolated islets were assessed using standard techniques. Results Leptin receptor mRNA levels were at or below the level of detection in wild-type adult mouse isolated islets and purified cells, and leptin signalling to Stat3 phosphorylation was undetectable. Whereas male mice further deleted for leptin receptors in β cells exhibited no abnormalities in glucose tolerance up to 16 weeks of age, females transiently displayed improved glucose tolerance at 8 weeks (11.2 ± 3.2% decrease in area under curve; p < 0.05), and improved (39.0 ± 13.0%, P < 0.05) glucose-stimulated insulin secretion in vitro. No differences were seen between genotypes in body weight, fasting glucose or β/α cell ratio. Deletion of LepR from α-cells, a minority of β cells, and a subset of proglucagon-expressing cells in the brain, exerted no effects on body weight, glucose or insulin tolerance, nor on pancreatic hormone secretion assessed in vivo and in vitro. Conclusions/interpretation The use here of a highly selective Cre recombinase indicates that leptin signalling plays a relatively minor, age- and sex-dependent role in the control of β cell function in the mouse. No in vivo role for leptin receptors on α cells, nor in other proglucagon-expressing cells, was detected in this study

    Leptin Production by Encapsulated Adipocytes Increases Brown Fat, Decreases Resistin, and Improves Glucose Intolerance in Obese Mice

    Get PDF
    <div><p>The neuroendocrine effects of leptin on metabolism hold promise to be translated into a complementary therapy to traditional insulin therapy for diabetes and obesity. However, injections of leptin can provoke inflammation. We tested the effects of leptin, produced in the physiological adipocyte location, on metabolism in mouse models of genetic and dietary obesity. We generated 3T3-L1 adipocytes constitutively secreting leptin and encapsulated them in a poly-L-lysine membrane, which protects the cells from immune rejection. <i>Ob/ob</i> mice (OB) were injected with capsules containing no cells (empty, OB<sup>[Emp]</sup>), adipocytes (OB<sup>[3T3]</sup>), or adipocytes overexpressing leptin (OB<sup>[<i>Lep</i>]</sup>) into both visceral fat depots. Leptin was found in the plasma of OB<sup>[<i>Lep</i>]</sup>, but not OB<sup>[Emp]</sup> and OB<sup>[3T3]</sup> mice at the end of treatment (72 days). The OB<sup>[<i>Lep</i>]</sup> and OB<sup>[3T3]</sup> mice have transiently suppressed appetite and weight loss compared to OB<sup>[Emp]</sup>. Only OB<sup>[<i>Lep</i>]</sup> mice have greater brown fat mass, metabolic rate, and reduced resistin plasma levels compared to OB<sup>[Emp]</sup>. Glucose tolerance was markedly better in OB<sup>[<i>Lep</i>]</sup> <i>vs</i>. OB<sup>[Emp]</sup> and OB<sup>[3T3]</sup> mice as well as in wild type mice with high-fat diet-induced obesity and insulin resistance treated with encapsulated leptin-producing adipocytes. Our proof-of-principle study provides evidence of long-term improvement of glucose tolerance with encapsulated adipocytes producing leptin.</p></div
    corecore