Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Endocrinology ; 161(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32267917

ABSTRACT

Targeted gene ablation studies of the endocrine pancreas have long suffered from suboptimal Cre deleter strains. In many cases, Cre lines purportedly specific for beta cells also displayed expression in other islet endocrine cells or in a subset of neurons in the brain. Several pancreas and endocrine Cre lines have experienced silencing or mosaicism over time. In addition, many Cre transgenic constructs were designed to include the hGH mini-gene, which by itself increases beta-cell replication and decreases beta-cell function. More recently, driver lines with Cre or CreER inserted into the Ins1 locus were generated, with the intent of producing ß cell-specific Cre lines with faithful recapitulation of insulin expression. These lines were bred in multiple labs to several different mouse lines harboring various lox alleles. In our hands, the ability of the Ins1-Cre and Ins1-CreER lines to delete target genes varied from that originally reported, with both alleles displaying low levels of expression, increased levels of methylation compared to the wild-type allele, and ultimately inefficient or absent target deletion. Thus, caution is warranted in the interpretation of results obtained with these genetic tools, and Cre expression and activity should be monitored regularly when using these lines.


Subject(s)
DNA Methylation/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Integrases/genetics , Recombination, Genetic/genetics , Alleles , Animals , Cells, Cultured , Female , Gene Silencing , HEK293 Cells , Humans , Insulin/metabolism , Insulin-Secreting Cells/physiology , Integrases/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics
2.
Nat Immunol ; 20(6): 677-686, 2019 06.
Article in English | MEDLINE | ID: mdl-31110312

ABSTRACT

Consumption of a high-energy Western diet triggers mild adaptive ß cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of ß cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of ß cells, but not that of α cells, leading to enlarged ß cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of ß cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse ß cell failure in patients with diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Proliferation , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Obesity/drug therapy , Parabiosis , Protein Binding , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
3.
J Gerontol A Biol Sci Med Sci ; 74(2): 155-162, 2019 01 16.
Article in English | MEDLINE | ID: mdl-29733330

ABSTRACT

Caloric restriction (CR) is the most potent nonpharmacological intervention known to both protect against carcinogenesis and delay aging in laboratory animals. There is a growing number of anticarcinogens and CR mimetics that activate NAD(P)H:quinone oxidoreductase 1 (NQO1). We have previously shown that NQO1, an antioxidant enzyme that acts as an energy sensor through modulation of intracellular redox and metabolic state, is upregulated by CR. Here, we used NQO1-knockout (KO) mice to investigate the role of NQO1 in both the aging process and tumor susceptibility, specifically in the context of CR. We found that NQO1 is not essential for the beneficial effects of CR on glucose homeostasis, physical performance, metabolic flexibility, life-span extension, and (unlike our previously observation with Nrf2) chemical-induced tumorigenesis.


Subject(s)
Body Composition , Caloric Restriction/adverse effects , Longevity , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms, Experimental/prevention & control , Oxidative Stress , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/etiology , Neoplasms, Experimental/metabolism
4.
Am J Physiol Endocrinol Metab ; 314(4): E308-E321, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351489

ABSTRACT

The transcription factors pancreatic and duodenal homeobox 1 (Pdx1) and onecut1 (Oc1) are coexpressed in multipotent pancreatic progenitors (MPCs), but their expression patterns diverge in hormone-expressing cells, with Oc1 expression being extinguished in the endocrine lineage and Pdx1 being maintained at high levels in ß-cells. We previously demonstrated that cooperative function of these two factors in MPCs is necessary for proper specification and differentiation of pancreatic endocrine cells. In those studies, we observed a persistent decrease in expression of the ß-cell maturity factor MafA. We therefore hypothesized that Pdx1 and Oc1 cooperativity in MPCs impacts postnatal ß-cell maturation and function. Here our model of Pdx1-Oc1 double heterozygosity was used to investigate the impact of haploinsufficiency for both of these factors on postnatal ß-cell maturation, function, and adaptability. Examining mice at postnatal day (P) 14, we observed alterations in pancreatic insulin content in both Pdx1 heterozygotes and double heterozygotes. Gene expression analysis at this age revealed significantly decreased expression of many genes important for glucose-stimulated insulin secretion (e.g., Glut2, Pcsk1/2, Abcc8) exclusively in double heterozygotes. Analysis of P14 islets revealed an increase in the number of mixed islets in double heterozygotes. We predicted that double-heterozygous ß-cells would have an impaired ability to respond to stress. Indeed, we observed that ß-cell proliferation fails to increase in double heterozygotes in response to either high-fat diet or placental lactogen. We thus report here the importance of cooperation between regulatory factors early in development for postnatal islet maturation and adaptability.


Subject(s)
Hepatocyte Nuclear Factor 6/physiology , Homeodomain Proteins/physiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/growth & development , Multipotent Stem Cells/metabolism , Trans-Activators/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Diet, High-Fat , Gene Expression Regulation, Developmental/drug effects , Glucose/pharmacology , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Male , Mice , Mice, Transgenic , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , Organogenesis/drug effects , Organogenesis/genetics , Trans-Activators/genetics
5.
Mol Metab ; 6(6): 548-559, 2017 06.
Article in English | MEDLINE | ID: mdl-28580285

ABSTRACT

OBJECTIVE: Hyperglycemia and systemic inflammation, hallmarks of Type 2 Diabetes (T2D), can induce the production of the inflammatory signaling molecule Prostaglandin E2 (PGE2) in islets. The effects of PGE2 are mediated by its four receptors, E-Prostanoid Receptors 1-4 (EP1-4). EP3 and EP4 play opposing roles in many cell types due to signaling through different G proteins, Gi and GS, respectively. We previously found that EP3 and EP4 expression are reciprocally regulated by activation of the FoxM1 transcription factor, which promotes ß-cell proliferation and survival. Our goal was to determine if EP3 and EP4 regulate ß-cell proliferation and survival and, if so, to elucidate the downstream signaling mechanisms. METHODS: ß-cell proliferation was assessed in mouse and human islets ex vivo treated with selective agonists and antagonists for EP3 (sulprostone and DG-041, respectively) and EP4 (CAY10598 and L-161,982, respectively). ß-cell survival was measured in mouse and human islets treated with the EP3- and EP4-selective ligands in conjunction with a cytokine cocktail to induce cell death. Changes in gene expression and protein phosphorylation were analyzed in response to modulation of EP3 and EP4 activity in mouse islets. RESULTS: Blockade of EP3 enhanced ß-cell proliferation in young, but not old, mouse islets in part through phospholipase C (PLC)-γ1 activity. Blocking EP3 also increased human ß-cell proliferation. EP4 modulation had no effect on ex vivo proliferation alone. However, blockade of EP3 in combination with activation of EP4 enhanced human, but not mouse, ß-cell proliferation. In both mouse and human islets, EP3 blockade or EP4 activation enhanced ß-cell survival in the presence of cytokines. EP4 acts in a protein kinase A (PKA)-dependent manner to increase mouse ß-cell survival. In addition, the positive effects of FoxM1 activation on ß-cell survival are inhibited by EP3 and dependent on EP4 signaling. CONCLUSIONS: Our results identify EP3 and EP4 as novel regulators of ß-cell proliferation and survival in mouse and human islets ex vivo.


Subject(s)
Cell Proliferation , Insulin-Secreting Cells/drug effects , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Acrylamides/pharmacology , Animals , Cell Survival , Cells, Cultured , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Phospholipase C gamma/metabolism , Protein Kinase C/metabolism , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Sulfones/pharmacology
6.
NPJ Aging Mech Dis ; 3: 8, 2017.
Article in English | MEDLINE | ID: mdl-28649426

ABSTRACT

Aging is a complex phenomenon involving functional decline in multiple physiological systems. We undertook a comparative analysis of skeletal muscle from four different species, i.e. mice, rats, rhesus monkeys, and humans, at three different representative stages during their lifespan (young, middle, and old) to identify pathways that modulate function and healthspan. Gene expression profiling and computational analysis revealed that pathway complexity increases from mice to humans, and as mammals age, there is predominantly an upregulation of pathways in all species. Two downregulated pathways, the electron transport chain and oxidative phosphorylation, were common among all four species in response to aging. Quantitative PCR, biochemical analysis, mitochondrial DNA measurements, and electron microscopy revealed a conserved age-dependent decrease in mitochondrial content, and a reduction in oxidative phosphorylation complexes in monkeys and humans. Western blot analysis of key proteins in mitochondrial biogenesis discovered that (i) an imbalance toward mitochondrial fusion occurs in aged skeletal muscle and (ii) mitophagy is not overtly affected, presumably leading to the observed accumulation of abnormally large, damaged mitochondria with age. Select transcript expression analysis uncovered that the skeletal inflammatory profile differentially increases with age, but is most pronounced in humans, while increased oxidative stress (as assessed by protein carbonyl adducts and 4-hydroxynonenal) is common among all species. Expression studies also found that there is unique dysregulation of the nutrient sensing pathways among the different species with age. The identification of conserved pathways indicates common molecular mechanisms intrinsic to health and lifespan, whereas the recognition of species-specific pathways emphasizes the importance of human studies for devising optimal therapeutic modalities to slow the aging process.

7.
J Cell Commun Signal ; 11(2): 105-116, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28132118

ABSTRACT

Prostaglandins (PGs) are signaling lipids derived from arachidonic acid (AA), which is metabolized by cyclooxygenase (COX)-1 or 2 and class-specific synthases to generate PGD2, PGE2, PGF2α, PGI2 (prostacyclin), and thromboxane A2. PGs signal through G-protein coupled receptors (GPCRs) and are important modulators of an array of physiological functions, including systemic inflammation and insulin secretion from pancreatic islets. The role of PGs in ß-cell function has been an active area of interest, beginning in the 1970s. Early studies demonstrated that PGE2 inhibits glucose-stimulated insulin secretion (GSIS), although more recent studies have questioned this inhibitory action of PGE2. The PGE2 receptor EP3 and one of the G-proteins that couples to EP3, GαZ, have been identified as negative regulators of ß-cell proliferation and survival. Conversely, PGI2 and its receptor, IP, play a positive role in the ß-cell by enhancing GSIS and preserving ß-cell mass in response to the ß-cell toxin streptozotocin (STZ). In comparison to PGE2 and PGI2, little is known about the function of the remaining PGs within islets. In this review, we discuss the roles of PGs, particularly PGE2 and PGI2, PG receptors, and downstream signaling events that alter ß-cell function and regulation of ß-cell mass.

8.
Physiol Rep ; 4(18)2016 Sep.
Article in English | MEDLINE | ID: mdl-27670405

ABSTRACT

Transgenic mouse models have been fundamental in the discovery of factors that regulate ß-cell development, mass, and function. Several groups have recently shown that some of these models display previously uncharacterized phenotypes due to the transgenic system itself. These include impaired islet function and increased ß-cell mass due to the presence of a human growth hormone (hGH) minigene as well as impaired ß-cell proliferation in response to tamoxifen (TM) administration. We aimed to determine how these systems impact ß-cell mass and proliferation during high fat diet (HFD). To this end, we utilized C57Bl6/J male MIP-CreER mice, which are known to express hGH, or wild-type (WT) mice treated with vehicle corn oil or TM In the absence of TM, MIP-CreER mice fed a chow diet have increased ß-cell mass due to hypertrophy, whereas replication is unchanged. Similarly, after 1 week on HFD, MIP-CreER mice have increased ß-cell mass compared to WT, and this is due to hypertrophy rather than increased proliferation. To assess the impact of TM on ß-cell proliferation and mass, WT mice were treated with vehicle corn oil or TM and then fed a chow diet or HFD for 3 days. We observed that TM-treated mice have improved glucose homeostasis on chow diet but impaired ß-cell proliferation in response to 3 days HFD feeding. These results unveil additional complications associated with commonly used pancreas-specific mouse models.

9.
Sci Rep ; 6: 28159, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27324831

ABSTRACT

The regulation of pancreatic ß cell mass is a critical factor to help maintain normoglycemia during insulin resistance. Nutrient-sensing G protein-coupled receptors (GPCR) contribute to aspects of ß cell function, including regulation of ß cell mass. Nutrients such as free fatty acids (FFAs) contribute to precise regulation of ß cell mass by signaling through cognate GPCRs, and considerable evidence suggests that circulating FFAs promote ß cell expansion by direct and indirect mechanisms. Free Fatty Acid Receptor 2 (FFA2) is a ß cell-expressed GPCR that is activated by short chain fatty acids, particularly acetate. Recent studies of FFA2 suggest that it may act as a regulator of ß cell function. Here, we set out to explore what role FFA2 may play in regulation of ß cell mass. Interestingly, Ffar2(-/-) mice exhibit diminished ß cell mass at birth and throughout adulthood, and increased ß cell death at adolescent time points, suggesting a role for FFA2 in establishment and maintenance of ß cell mass. Additionally, activation of FFA2 with Gαq/11-biased agonists substantially increased ß cell proliferation in in vitro and ex vivo proliferation assays. Collectively, these data suggest that FFA2 may be a novel therapeutic target to stimulate ß cell growth and proliferation.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/pathology , Receptors, Cell Surface/metabolism , Animals , Cell Survival , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Volatile/metabolism , Humans , Insulin Resistance , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Signal Transduction
10.
Endocrinology ; 157(1): 220-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26485614

ABSTRACT

Mice carrying a targeted disruption of the prostaglandin E2 (PGE2) E-prostanoid receptor 3 (EP3) gene, Ptger3, were fed a high-fat diet (HFD), or a micronutrient matched control diet, to investigate the effects of disrupted PGE2-EP3 signaling on diabetes in a setting of diet-induced obesity. Although no differences in body weight were seen in mice fed the control diet, when fed a HFD, EP3(-/-) mice gained more weight relative to EP3(+/+) mice. Overall, EP3(-/-) mice had increased epididymal fat mass and adipocyte size; paradoxically, a relative decrease in both epididymal fat pad mass and adipocyte size was observed in the heaviest EP3(-/-) mice. The EP3(-/-) mice had increased macrophage infiltration, TNF-α, monocyte chemoattractant protein-1, IL-6 expression, and necrosis in their epididymal fat pads as compared with EP3(+/+) animals. Adipocytes isolated from EP3(+/+) or EP3(-/-) mice were assayed for the effect of PGE2-evoked inhibition of lipolysis. Adipocytes isolated from EP3(-/-) mice lacked PGE2-evoked inhibition of isoproterenol stimulated lipolysis compared with EP3(+/+). EP3(-/-) mice fed HFD had exaggerated ectopic lipid accumulation in skeletal muscle and liver, with evidence of hepatic steatosis. Both blood glucose and plasma insulin levels were similar between genotypes on a control diet, but when fed HFD, EP3(-/-) mice became hyperglycemic and hyperinsulinemic when compared with EP3(+/+) fed HFD, demonstrating a more severe insulin resistance phenotype in EP3(-/-). These results demonstrate that when fed a HFD, EP3(-/-) mice have abnormal lipid distribution, developing excessive ectopic lipid accumulation and associated insulin resistance.


Subject(s)
Adipose Tissue, White/metabolism , Adiposity , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat/adverse effects , Obesity/metabolism , Panniculitis/etiology , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/pathology , Animals , Cell Size , Crosses, Genetic , Diabetes Mellitus, Type 2/immunology , Insulin Resistance , Lipid Metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophage Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Necrosis , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Obesity/etiology , Obesity/pathology , Obesity/physiopathology , Panniculitis/immunology , Receptors, Prostaglandin E, EP3 Subtype/genetics , Weight Gain
11.
Am J Physiol Endocrinol Metab ; 308(7): E573-82, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25628421

ABSTRACT

Both short- (1 wk) and long-term (2-12 mo) high-fat diet (HFD) studies reveal enhanced ß-cell mass due to increased ß-cell proliferation. ß-Cell proliferation following HFD has been postulated to occur in response to insulin resistance; however, whether HFD can induce ß-cell proliferation independent of insulin resistance has been controversial. To examine the kinetics of HFD-induced ß-cell proliferation and its correlation with insulin resistance, we placed 8-wk-old male C57Bl/6J mice on HFD for different lengths of time and assayed the following: glucose tolerance, insulin secretion in response to glucose, insulin tolerance, ß-cell mass, and ß-cell proliferation. We found that ß-cell proliferation was significantly increased after only 3 days of HFD feeding, weeks before an increase in ß-cell mass or peripheral insulin resistance was detected. These results were confirmed by hyperinsulinemic euglycemic clamps and measurements of α-hydroxybutyrate, a plasma biomarker of insulin resistance in humans. An increase in expression of key islet-proliferative genes was found in isolated islets from 1-wk HFD-fed mice compared with chow diet (CD)-fed mice. These data indicate that short-term HFD feeding enhances ß-cell proliferation before insulin resistance becomes apparent.


Subject(s)
Cell Proliferation , Diet, High-Fat , Insulin Resistance , Insulin-Secreting Cells/physiology , Animals , Cell Proliferation/drug effects , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Glucose Clamp Technique , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Time Factors
12.
Ageing Res Rev ; 11(3): 390-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22210414

ABSTRACT

During aging there is an increasing imbalance of energy intake and expenditure resulting in obesity, frailty, and metabolic disorders. For decades, research has shown that caloric restriction (CR) and exercise can postpone detrimental aspects of aging. These two interventions invoke a similar physiological signature involving pathways associated with stress responses and mitochondrial homeostasis. Nonetheless, CR is able to delay aging processes that result in an increase of both mean and maximum lifespan, whereas exercise primarily increases healthspan. Due to the strict dietary regime necessary to achieve the beneficial effects of CR, most studies to date have focused on rodents and non-human primates. As a consequence, there is vast interest in the development of compounds such as resveratrol, metformin and rapamycin that would activate the same metabolic- and stress-response pathways induced by these interventions without actually restricting caloric intake. Therefore the scope of this review is to (i) describe the benefits of CR and exercise in healthy individuals, (ii) discuss the role of these interventions in the diseased state, and (iii) examine some of the promising pharmacological alternatives such as CR- and exercise-mimetics.


Subject(s)
Aging/physiology , Biomimetic Materials/metabolism , Caloric Restriction , Exercise/physiology , Animals , Biomimetic Materials/therapeutic use , Caloric Restriction/methods , Energy Intake/physiology , Humans , Longevity/physiology , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...