43 results on '"Gareth E, Lim"'
Search Results
2. Identification of biomarkers for glycaemic deterioration in type 2 diabetes
- Author
-
Roderick C. Slieker, Louise A. Donnelly, Elina Akalestou, Livia Lopez-Noriega, Rana Melhem, Ayşim Güneş, Frederic Abou Azar, Alexander Efanov, Eleni Georgiadou, Hermine Muniangi-Muhitu, Mahsa Sheikh, Giuseppe N. Giordano, Mikael Åkerlund, Emma Ahlqvist, Ashfaq Ali, Karina Banasik, Søren Brunak, Marko Barovic, Gerard A. Bouland, Frédéric Burdet, Mickaël Canouil, Iulian Dragan, Petra J. M. Elders, Celine Fernandez, Andreas Festa, Hugo Fitipaldi, Phillippe Froguel, Valborg Gudmundsdottir, Vilmundur Gudnason, Mathias J. Gerl, Amber A. van der Heijden, Lori L. Jennings, Michael K. Hansen, Min Kim, Isabelle Leclerc, Christian Klose, Dmitry Kuznetsov, Dina Mansour Aly, Florence Mehl, Diana Marek, Olle Melander, Anne Niknejad, Filip Ottosson, Imre Pavo, Kevin Duffin, Samreen K. Syed, Janice L. Shaw, Over Cabrera, Timothy J. Pullen, Kai Simons, Michele Solimena, Tommi Suvitaival, Asger Wretlind, Peter Rossing, Valeriya Lyssenko, Cristina Legido Quigley, Leif Groop, Bernard Thorens, Paul W. Franks, Gareth E. Lim, Jennifer Estall, Mark Ibberson, Joline W. J. Beulens, Leen M ’t Hart, Ewan R. Pearson, and Guy A. Rutter
- Subjects
Science - Abstract
Abstract We identify biomarkers for disease progression in three type 2 diabetes cohorts encompassing 2,973 individuals across three molecular classes, metabolites, lipids and proteins. Homocitrulline, isoleucine and 2-aminoadipic acid, eight triacylglycerol species, and lowered sphingomyelin 42:2;2 levels are predictive of faster progression towards insulin requirement. Of ~1,300 proteins examined in two cohorts, levels of GDF15/MIC-1, IL-18Ra, CRELD1, NogoR, FAS, and ENPP7 are associated with faster progression, whilst SMAC/DIABLO, SPOCK1 and HEMK2 predict lower progression rates. In an external replication, proteins and lipids are associated with diabetes incidence and prevalence. NogoR/RTN4R injection improved glucose tolerance in high fat-fed male mice but impaired it in male db/db mice. High NogoR levels led to islet cell apoptosis, and IL-18R antagonised inflammatory IL-18 signalling towards nuclear factor kappa-B in vitro. This comprehensive, multi-disciplinary approach thus identifies biomarkers with potential prognostic utility, provides evidence for possible disease mechanisms, and identifies potential therapeutic avenues to slow diabetes progression.
- Published
- 2023
- Full Text
- View/download PDF
3. 14-3-3ζ Constrains insulin secretion by regulating mitochondrial function in pancreatic β cells
- Author
-
Yves Mugabo, Cheng Zhao, Ju Jing Tan, Anindya Ghosh, Scott A. Campbell, Evgenia Fadzeyeva, Frédéric Paré, Siew Siew Pan, Maria Galipeau, Julia Ast, Johannes Broichhagen, David J. Hodson, Erin E. Mulvihill, Sophie Petropoulos, and Gareth E. Lim
- Subjects
Endocrinology ,Metabolism ,Medicine - Abstract
While critical for neurotransmitter synthesis, 14-3-3 proteins are often assumed to have redundant functions due to their ubiquitous expression, but despite this assumption, various 14-3-3 isoforms have been implicated in regulating metabolism. We previously reported contributions of 14-3-3ζ in β cell function, but these studies were performed in tumor-derived MIN6 cells and systemic KO mice. To further characterize the regulatory roles of 14-3-3ζ in β cell function, we generated β cell–specific 14-3-3ζ–KO mice. Although no effects on β cell mass were detected, potentiated glucose-stimulated insulin secretion (GSIS), mitochondrial function, and ATP synthesis were observed. Deletion of 14-3-3ζ also altered the β cell transcriptome, as genes associated with mitochondrial respiration and oxidative phosphorylation were upregulated. Acute 14-3-3 protein inhibition in mouse and human islets recapitulated the enhancements in GSIS and mitochondrial function, suggesting that 14-3-3ζ is the critical isoform in β cells. In dysfunctional db/db islets and human islets from type 2 diabetic donors, expression of Ywhaz/YWHAZ, the gene encoding 14-3-3ζ, was inversely associated with insulin secretion, and pan–14-3-3 protein inhibition led to enhanced GSIS and mitochondrial function. Taken together, this study demonstrates important regulatory functions of 14-3-3ζ in the regulation of β cell function and provides a deeper understanding of how insulin secretion is controlled in β cells.
- Published
- 2022
- Full Text
- View/download PDF
4. Metabolic Contributions of Wnt Signaling: More Than Controlling Flight
- Author
-
Frederic Abou Azar and Gareth E. Lim
- Subjects
Wnt signaling ,β-catenin ,TCF7L2 ,GSK-3β ,metabolism ,embryogenesis ,Biology (General) ,QH301-705.5 - Abstract
The canonical Wnt signaling pathway is ubiquitous throughout the body and influences a diverse array of physiological processes. Following the initial discovery of the Wnt signaling pathway during wing development in Drosophila melanogaster, it is now widely appreciated that active Wnt signaling in mammals is necessary for the development and growth of various tissues involved in whole-body metabolism, such as brain, liver, pancreas, muscle, and adipose. Moreover, elegant gain- and loss-of-function studies have dissected the tissue-specific roles of various downstream effector molecules in the regulation of energy homeostasis. This review attempts to highlight and summarize the contributions of the Wnt signaling pathway and its downstream effectors on whole-body metabolism and their influence on the development of metabolic diseases, such as diabetes and obesity. A better understanding of the Wnt signaling pathway in these tissues may aid in guiding the development of future therapeutics to treat metabolic diseases.
- Published
- 2021
- Full Text
- View/download PDF
5. 14-3-3ζ mediates an alternative, non-thermogenic mechanism in male mice to reduce heat loss and improve cold tolerance
- Author
-
Kadidia Diallo, Sylvie Dussault, Christophe Noll, Angel F. Lopez, Alain Rivard, André C. Carpentier, and Gareth E. Lim
- Subjects
14-3-3 proteins ,14-3-3ζ ,Beiging ,Vasoconstriction ,Adaptive thermogenesis ,Internal medicine ,RC31-1245 - Abstract
Objective: Adaptive thermogenesis, which is partly mediated by sympathetic input on brown adipose tissue (BAT), is a mechanism of heat production that confers protection against prolonged cold exposure. Various endogenous stimuli, for example, norepinephrine and FGF-21, can also promote the conversion of inguinal white adipocytes to beige adipocytes, which may represent a secondary cell type that contributes to adaptive thermogenesis. We previously identified an essential role of the molecular scaffold 14-3-3ζ in adipogenesis, but one of the earliest, identified functions of 14-3-3ζ is its regulatory effects on the activity of tyrosine hydroxylase, the rate-limiting enzyme in the synthesis of norepinephrine. Herein, we examined whether 14-3-3ζ could influence adaptive thermogenesis via actions on BAT activation or the beiging of white adipocytes. Methods: Transgenic mice over-expressing a TAP-tagged human 14-3-3ζ molecule or heterozygous mice without one allele of Ywhaz, the gene encoding 14-3-3ζ, were used to explore the contribution of 14-3-3ζ to acute (3 h) and prolonged (3 days) cold (4 °C) exposure. Metabolic caging experiments, PET-CT imaging, and laser Doppler imaging were used to determine the effect of 14-3-3ζ over-expression on thermogenic and vasoconstrictive mechanisms in response to cold. Results: Transgenic over-expression of 14-3-3ζ (TAP) in male mice significantly improved tolerance to acute and prolonged cold. In response to cold, body temperatures in TAP mice did not decrease to the same extent when compared to wildtype (WT) mice, and this was associated with increased UCP1 expression in beige inguinal white tissue (iWAT) and BAT. Of note was the paradoxical finding that cold-induced changes in body temperatures of TAP mice were associated with significantly decreased energy expenditure. The marked improvements in tolerance to prolonged cold were not due to changes in sensitivity to β-adrenergic stimulation or BAT or iWAT oxidative metabolism; instead, over-expression of 14-3-3ζ significantly decreased thermal conductance and heat loss in mice via increased peripheral vasoconstriction. Conclusions: Despite being associated with elevations in cold-induced UCP1 expression in brown or beige adipocytes, these findings suggest that 14-3-3ζ regulates an alternative, non-thermogenic mechanism via vasoconstriction to minimize heat loss during cold exposure.
- Published
- 2020
- Full Text
- View/download PDF
6. Reduced Circulating Insulin Enhances Insulin Sensitivity in Old Mice and Extends Lifespan
- Author
-
Nicole M. Templeman, Stephane Flibotte, Jenny H.L. Chik, Sunita Sinha, Gareth E. Lim, Leonard J. Foster, Corey Nislow, and James D. Johnson
- Subjects
hyperinsulinemia ,insulin resistance ,longevity ,aging ,metabolism ,Biology (General) ,QH301-705.5 - Abstract
The causal relationships between insulin levels, insulin resistance, and longevity are not fully elucidated. Genetic downregulation of insulin/insulin-like growth factor 1 (Igf1) signaling components can extend invertebrate and mammalian lifespan, but insulin resistance, a natural form of decreased insulin signaling, is associated with greater risk of age-related disease in mammals. We compared Ins2+/− mice to Ins2+/+ littermate controls, on a genetically stable Ins1 null background. Proteomic and transcriptomic analyses of livers from 25-week-old mice suggested potential for healthier aging and altered insulin sensitivity in Ins2+/− mice. Halving Ins2 lowered circulating insulin by 25%–34% in aged female mice, without altering Igf1 or circulating Igf1. Remarkably, decreased insulin led to lower fasting glucose and improved insulin sensitivity in aged mice. Moreover, lowered insulin caused significant lifespan extension, observed across two diverse diets. Our study indicates that elevated insulin contributes to age-dependent insulin resistance and that limiting basal insulin levels can extend lifespan.
- Published
- 2017
- Full Text
- View/download PDF
7. Inter-domain tagging implicates caveolin-1 in insulin receptor trafficking and Erk signaling bias in pancreatic beta-cells
- Author
-
Tobias Boothe, Gareth E. Lim, Haoning Cen, Søs Skovsø, Micah Piske, Shu Nan Li, Ivan R. Nabi, Patrick Gilon, and James D. Johnson
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: The role and mechanisms of insulin receptor internalization remain incompletely understood. Previous trafficking studies of insulin receptors involved fluorescent protein tagging at their termini, manipulations that may be expected to result in dysfunctional receptors. Our objective was to determine the trafficking route and molecular mechanisms of functional tagged insulin receptors and endogenous insulin receptors in pancreatic beta-cells. Methods: We generated functional insulin receptors tagged with pH-resistant fluorescent proteins between domains. Confocal, TIRF and STED imaging revealed a trafficking pattern of inter-domain tagged insulin receptors and endogenous insulin receptors detected with antibodies. Results: Surprisingly, interdomain-tagged and endogenous insulin receptors in beta-cells bypassed classical Rab5a- or Rab7-mediated endocytic routes. Instead, we found that removal of insulin receptors from the plasma membrane involved tyrosine-phosphorylated caveolin-1, prior to trafficking within flotillin-1-positive structures to lysosomes. Multiple methods of inhibiting caveolin-1 significantly reduced Erk activation in vitro or in vivo, while leaving Akt signaling mostly intact. Conclusions: We conclude that phosphorylated caveolin-1 plays a role in insulin receptor internalization towards lysosomes through flotillin-1-positive structures and that caveolin-1 helps bias physiological beta-cell insulin signaling towards Erk activation. Author Video: Author Video Watch what authors say about their articles Keywords: Insulin receptor internalization, Insulin resistance, Pancreatic islet beta-cells, Autocrine insulin signaling
- Published
- 2016
- Full Text
- View/download PDF
8. 1396-P: Critical Cellular Functions That Mediate the Early Stages of Adipogenesis Are Directly Influenced by 14-3-3zeta and Its Interactome
- Author
-
SABRI A. RIAL, AMAL ALKHOURY, GENEVIEVE LAVOIE, PHILIPPE ROUX, THOMAS M. DURCAN, AIDA MARTINEZ-SANCHEZ, and GARETH E. LIM
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Abstract
Aim: We previously discovered that 14-3-3-zeta (14-3-3ζ) was essential for adipocyte differentiation. Although 14-3-3ζ scaffold is not a transcription factor (TF) , it undergoes nuclear translocation during adipogenesis. Considering its diverse interactome, we hypothesized that 14-3-3ζ anchors, nucleates and regulates essential transcriptional complexes required for adipogenesis. Methods: 3T3-L1 pre-adipocytes were modified by CRISPR-Cas9 to express a TAP (Flag-HA) -tagged 14-3-3ζ protein. At 48 first hours of adipogenesis, nuclear TAP-14-3-3-ζ complexes were purified to identify and annotate the 14-3-3ζ interactome by MS and Gene Ontology. Highly enriched proteins functions were assessed for their roles in adipogenesis using pharmacological inhibitors or siRNA. In parallel, chromatin accessibility was assessed by ATAC-Seq from control or 14-3-3ζ-depleted 3T3-L1 cells during early adipogenesis. Results: We found that the 14-3-3ζ nuclear interactome during the early stages of adipogenesis was significantly enriched with 133 proteins, including CEBP-β, an early adipogenic TF. According to GO analysis, proteins within the 14-3-3ζ interactome were primarily associated (FDR ≤ 0.01) with histone H2B ubiquitination, DNA unwinding for replication, DNA hypermethylation and ribosome assembly. Pharmacological inhibition or depletion by siRNA of these proteins significantly dampened adipocyte differentiation. Principal component analysis of ATAC-seq results revealed that samples from 14-3-3ζ-depleted cells differed from controls in term of whole chromatin accessibility, and DESeq2 identified nearly 3400 (p Conclusion: These results reveal that via critical protein interactions, 14-3-3ζ has essential roles in chromatin accessibility, DNA replication, and mRNA translation, which are crucial events for the early adipogenesis. Disclosure S.A.Rial: None. A.Alkhoury: None. G.Lavoie: None. P.Roux: None. T.M.Durcan: None. A.Martinez-sanchez: None. G.E.Lim: None. Funding Natural Sciences and Engineering Research Council of Canada (NSERC) ; Canada Research Chairs (CRC) ; Fonds de recherche du Québec-Nature and Technologies (FRQNT)
- Published
- 2022
- Full Text
- View/download PDF
9. The Application of High‐Throughput Approaches in Identifying Novel Therapeutic Targets and Agents to Treat Diabetes
- Author
-
Siyi He and Gareth E. Lim
- Subjects
Biomaterials ,Biomedical Engineering ,General Biochemistry, Genetics and Molecular Biology - Abstract
During the past decades, unprecedented progress in technologies has revolutionized traditional research methodologies. Among these, advances in high-throughput drug screening approaches have permitted the rapid identification of potential therapeutic agents from drug libraries that contain thousands or millions of molecules. Moreover, high-throughput-based therapeutic target discovery strategies can comprehensively interrogate relationships between biomolecules (e.g., gene, RNA, and protein) and diseases and significantly increase the authors' knowledge of disease mechanisms. Diabetes is a chronic disease primarily characterized by the incapacity of the body to maintain normoglycemia. The prevalence of diabetes in modern society has become a severe public health issue that threatens the well-being of millions of patients. Although a number of pharmacological treatments are available, there is no permanent cure for diabetes, and discovering novel therapeutic targets and agents continues to be an urgent need. The present review discusses the technical details of high-throughput screening approaches in drug discovery, followed by introducing the applications of such approaches to diabetes research. This review aims to provide an example of the applicability of high-throughput technologies in facilitating different aspects of disease research.
- Published
- 2022
- Full Text
- View/download PDF
10. Is dynamic autocrine insulin signaling possible? A mathematical model predicts picomolar concentrations of extracellular monomeric insulin within human pancreatic islets.
- Author
-
Minghu Wang, Jiaxu Li, Gareth E Lim, and James D Johnson
- Subjects
Medicine ,Science - Abstract
Insulin signaling is essential for β-cell survival and proliferation in vivo. Insulin also has potent mitogenic and anti-apoptotic actions on cultured β-cells, with maximum effect in the high picomolar range and diminishing effect at high nanomolar doses. In order to understand whether these effects of insulin are constitutive or can be subjected to physiological modulation, it is essential to estimate the extracellular concentration of monomeric insulin within an intact islet. Unfortunately, the in vivo concentration of insulin monomers within the islet cannot be measured directly with current technology. Here, we present the first mathematical model designed to estimate the levels of monomeric insulin within the islet extracellular space. Insulin is released as insoluble crystals that exhibit a delayed dissociation into hexamers, dimers, and eventually monomers, which only then can act as signaling ligands. The rates at which different forms of insulin dissolve in vivo have been estimated from studies of peripheral insulin injection sites. We used this and other information to formulate a mathematical model to estimate the local insulin concentration within a single islet as a function of glucose. Model parameters were estimated from existing literature. Components of the model were validated using experimental data, if available. Model analysis predicted that the majority of monomeric insulin in the islet is that which has been returned from the periphery, and the concentration of intra-islet monomeric insulin varies from ~50-300 pM when glucose is in the physiological range. Thus, our results suggest that the local concentration of monomeric insulin within the islet is in the picomolar 'sweet spot' range of insulin doses that activate the insulin receptor and have the most potent effects on β-cells in vitro. Together with experimental data, these estimations support the concept that autocrine/paracrine insulin signalling within the islet is dynamic, rather than constitutive and saturated.
- Published
- 2013
- Full Text
- View/download PDF
11. 14-3-3ζ mediates an alternative, non-thermogenic mechanism in male mice to reduce heat loss and improve cold tolerance
- Author
-
Angel F. Lopez, André C. Carpentier, Gareth E. Lim, Alain Rivard, Kadidia Diallo, Sylvie Dussault, Christophe Noll, Diallo, Kadidia, Dussault, Sylvie, Noll, Christophe, Lopez, Angel F, Rivard, Alain, Carpentier, André C, and Lim, Gareth E
- Subjects
0301 basic medicine ,Male ,adaptive thermogenesis ,Stimulation ,Mice ,0302 clinical medicine ,Adipose Tissue, Brown ,Positron Emission Tomography Computed Tomography ,Brown adipose tissue ,vasoconstriction ,Uncoupling Protein 1 ,Adipogenesis ,Chemistry ,Thermogenesis ,Cold Temperature ,medicine.anatomical_structure ,14-3-3z ,YWHAZ ,Original Article ,medicine.symptom ,Body Temperature Regulation ,Genetically modified mouse ,Thermotolerance ,medicine.medical_specialty ,lcsh:Internal medicine ,Transgene ,Adipose Tissue, White ,030209 endocrinology & metabolism ,Mice, Transgenic ,14-3-3ζ ,Mitochondrial Proteins ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,lcsh:RC31-1245 ,Molecular Biology ,Tyrosine hydroxylase ,Cold-Shock Response ,Beiging ,14-3-3 proteins ,Cell Biology ,Adaptive thermogenesis ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Vasoconstriction ,beiging - Abstract
Objective Adaptive thermogenesis, which is partly mediated by sympathetic input on brown adipose tissue (BAT), is a mechanism of heat production that confers protection against prolonged cold exposure. Various endogenous stimuli, for example, norepinephrine and FGF-21, can also promote the conversion of inguinal white adipocytes to beige adipocytes, which may represent a secondary cell type that contributes to adaptive thermogenesis. We previously identified an essential role of the molecular scaffold 14-3-3ζ in adipogenesis, but one of the earliest, identified functions of 14-3-3ζ is its regulatory effects on the activity of tyrosine hydroxylase, the rate-limiting enzyme in the synthesis of norepinephrine. Herein, we examined whether 14-3-3ζ could influence adaptive thermogenesis via actions on BAT activation or the beiging of white adipocytes. Methods Transgenic mice over-expressing a TAP-tagged human 14-3-3ζ molecule or heterozygous mice without one allele of Ywhaz, the gene encoding 14-3-3ζ, were used to explore the contribution of 14-3-3ζ to acute (3 h) and prolonged (3 days) cold (4 °C) exposure. Metabolic caging experiments, PET-CT imaging, and laser Doppler imaging were used to determine the effect of 14-3-3ζ over-expression on thermogenic and vasoconstrictive mechanisms in response to cold. Results Transgenic over-expression of 14-3-3ζ (TAP) in male mice significantly improved tolerance to acute and prolonged cold. In response to cold, body temperatures in TAP mice did not decrease to the same extent when compared to wildtype (WT) mice, and this was associated with increased UCP1 expression in beige inguinal white tissue (iWAT) and BAT. Of note was the paradoxical finding that cold-induced changes in body temperatures of TAP mice were associated with significantly decreased energy expenditure. The marked improvements in tolerance to prolonged cold were not due to changes in sensitivity to β-adrenergic stimulation or BAT or iWAT oxidative metabolism; instead, over-expression of 14-3-3ζ significantly decreased thermal conductance and heat loss in mice via increased peripheral vasoconstriction. Conclusions Despite being associated with elevations in cold-induced UCP1 expression in brown or beige adipocytes, these findings suggest that 14-3-3ζ regulates an alternative, non-thermogenic mechanism via vasoconstriction to minimize heat loss during cold exposure., Graphical abstract Image 1, Highlights • 14-3-3ζ over-expression in male mice improves tolerance to acute and prolonged cold. • Increasing 14-3-3ζ expression promotes beiging of inguinal white adipose tissue. • Cold-induced changes in body temperature can be dissociated from energy expenditure. • 14-3-3ζ-dependent decreases in heat loss are associated with vasoconstriction.
- Published
- 2020
12. 321-OR: Deletion of 14-3-3ζ in Pancreatic ß-Cells Increase Mitochondrial Activity and Insulin Secretion
- Author
-
Yves Mugabo, Ryszard Grygorczyk, Gareth E. Lim, Ju Jing Tan, Maria Galipeau, and Evgenia Fadzeyeva
- Subjects
geography ,medicine.medical_specialty ,geography.geographical_feature_category ,ATP synthase ,biology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Islet ,Calcium in biology ,Endocrinology ,Internal medicine ,Knockout mouse ,Internal Medicine ,biology.protein ,medicine ,Glucose homeostasis ,Glycolysis ,Secretion - Abstract
Pancreatic β-cells continuously sense blood glucose levels and secrete insulin to maintain normoglycemia. In β-cells, ATP generated by glycolysis promotes the closure of ATP-sensitive K+ channels, thereby increasing intracellular calcium and ultimately insulin secretion. 14-3-3 proteins, and in particular 14-3-3ζ, have been found to regulate ATP synthase and mitochondrial respiration, suggesting that members of this family of scaffold proteins in β-cells may influence glucose-stimulated insulin secretion (GSIS) and glucose homeostasis. To date, we have identified critical contributions of 14-3-3 proteins to GSIS. In mouse and human islets, pan-inhibition of 14-3-3 proteins with cell-permeable inhibitors potentiated ex-vivo GSIS. This was associated with increased mitochondrial function, as oxygen consumption and ATP synthesis rates were significantly enhanced. Moreover, increased ATP production was confirmed with luciferase-based assays. Of the seven isoforms, we previously reported critical metabolic roles of 14-3-3ζ in glucose homeostasis, and to understand its role in β-cells, β-cell-specific knockout mice (Ins1CreThor:14-3-3ζfl/fl, β-KO) were generated. When compared to control mice, no differences in body weights or glucose and insulin tolerance were observed, but β-KO mice displayed significantly enhanced insulin secretion following i.p. glucose (2 g/kg), and ex vivo islet perifusion studies revealed enhanced 2nd-phase secretion of GSIS from β-KO islets. Similar to pan-14-3-3 inhibition, increased mitochondrial activity and ATP synthesis were detected in islets of β-KO mice. In summary, these results demonstrate critical functions of 14-3-3ζ and its related proteins in mitochondrial activity in β-cells and the regulation of GSIS. These data also suggest that 14-3-3ζ inhibition may represent a promising target to enhance pancreatic β-cell function in the context of diabetes. Disclosure Y. Mugabo: None. M. Galipeau: None. J. Tan: None. E. Fadzeyeva: None. R. Grygorczyk: None. G.E. Lim: None. Funding Canadian Institutes of Health Research; Canada Research Chairs Program (PJT-153144)
- Published
- 2020
- Full Text
- View/download PDF
13. Reducing 14-3-3ζ expression influences adipocyte maturity and impairs function
- Author
-
Abel Oppong, Christine Des Rosiers, Kadidia Diallo, Gareth E. Lim, and Isabelle Robillard Frayne
- Subjects
0301 basic medicine ,Glycerol ,Male ,medicine.medical_specialty ,Physiology ,Endocrinology, Diabetes and Metabolism ,Lipolysis ,Peroxisome proliferator-activated receptor ,Adipose tissue ,Plasma protein binding ,Fatty Acids, Nonesterified ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Adipocyte ,3T3-L1 Cells ,Lipidomics ,medicine ,Adipocytes ,Animals ,RNA, Messenger ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Adipogenesis ,Catabolism ,Cell Differentiation ,Lipase ,Peroxisome ,Sterol Esterase ,Cell biology ,PPAR gamma ,030104 developmental biology ,Endocrinology ,chemistry ,14-3-3 Proteins ,030220 oncology & carcinogenesis ,Phosphorylation - Abstract
One of the primary metabolic functions of a mature adipocyte is to supply energy via lipolysis, or the catabolism of stored lipids. Hormone-sensitive lipase (HSL) is a critical lipolytic enzyme, and its phosphorylation and subsequent activation by PKA generates phospho-binding sites for 14-3-3 proteins, a ubiquitously expressed family of molecular scaffolds. While we previously identified essential roles of the 14-3-3ζ isoform in murine adipogenesis, the presence of 14-3-3 protein binding sites on HSL suggests that 14-3-3ζ could also influence mature adipocyte processes like lipolysis. Herein, we demonstrate that 14-3-3ζ is necessary for lipolysis in male mice and fully differentiated 3T3-L1 adipocytes, as depletion of 14-3-3ζ significantly impaired glycerol and FFA release. Unexpectedly, this was not due to impairments in signaling events underlying lipolysis; instead, reducing 14-3-3ζ expression was found to significantly impact adipocyte maturity, as observed by reduced abundance of PPARγ2 protein and expression of mature adipocytes genes and those associated withde novotriglyceride synthesis and lipolysis. The impact of 14-3-3ζ depletion on adipocyte maturity was further examined with untargeted lipidomics, which revealed that reductions in 14-3-3ζ abundance promoted the acquisition of a lipidomic signature that resembled undifferentiated, pre-adipocytes. Collectively, these findings reveal a novel aspect of 14-3-3ζ in adipocytes, as reducing 14-3-3ζ was found to have a negative effect on adipocyte maturity and adipocyte-specific processes like lipolysis.
- Published
- 2020
14. Can 14-3-3 proteins serve as therapeutic targets for the treatment of metabolic diseases?
- Author
-
Kadidia Diallo, Gareth E. Lim, and Abel Oppong
- Subjects
0301 basic medicine ,Pharmacology ,Scaffold protein ,chemistry.chemical_classification ,Protein family ,Cancer ,Computational biology ,Metabolism ,Biology ,medicine.disease ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Enzyme ,14-3-3 Proteins ,Metabolic Diseases ,chemistry ,030220 oncology & carcinogenesis ,Diabetes mellitus ,medicine ,Animals ,Humans ,Obesity ,Serotonin biosynthesis - Abstract
Since their initial characterization as abundant brain proteins more than 5 decades ago, a resurgence into understanding the cellular functions of 14-3-3 proteins has emerged. While one of the earliest functions attributed to this eukaryotic scaffold protein family was the activation of enzymes involved in catecholamine and serotonin biosynthesis, 14-3-3 proteins have since been implicated in the regulation of several cellular processes including cell-cycle control, apoptosis, and metabolism. Moreover, increasing lines of evidence demonstrate links between changes in 14-3-3 protein function and the pathogenesis of chronic diseases. As a result, this has raised the question of whether 14-3-3 proteins represent viable targets for pharmacological intervention against diseases such as obesity, diabetes and cancer. In addition to providing an overview of the 14-3-3 protein family, we will discuss their connections to metabolism and metabolic diseases. We will also elaborate on the potential of targeting 14-3-3 proteins, as well as components of their interactomes, for developing novel therapies for treating metabolic diseases, including diabetes and obesity.
- Published
- 2019
- Full Text
- View/download PDF
15. Adipose depot-specific upregulation of Ucp1 or mitochondrial oxidative complex proteins are early consequences of genetic insulin reduction in mice
- Author
-
James D. Johnson, Obelia Haida, Peter Overby, Gareth E. Lim, José Rodrigo Pauli, Nicole M. Templeman, Wang S, José Diego Botezelli, and Lorenzo Lindo
- Subjects
endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Fasting hyperinsulinemia ,medicine.medical_treatment ,030209 endocrinology & metabolism ,White adipose tissue ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Adipocyte ,Brown adipose tissue ,medicine ,Hyperinsulinemia ,030304 developmental biology ,2. Zero hunger ,PRDM16 ,0303 health sciences ,Chemistry ,Insulin ,food and beverages ,nutritional and metabolic diseases ,medicine.disease ,Thermogenin ,medicine.anatomical_structure ,Endocrinology ,hormones, hormone substitutes, and hormone antagonists - Abstract
Hyperinsulinemia plays a causal role in adiposity tissue expansion. We have previously shown that mice with reduced insulin gene dosage have increased energy expenditure, but the tissue-specific molecular mechanisms involved in the effects of abrogated hyperinsulinemia have remained unclear. Herein we investigated the effects of genetically reducing insulin production on the abundance of oxidative mitochondrial complex proteins in liver, skeletal muscle, white adipose tissue and brown adipose tissue. To suppress insulin levels, we manipulated Ins1 gene dosage in mice lacking both Ins2 alleles to prevent compensation. Male Ins1+/+ or Ins1+/- littermates were fed either a low-fat diet (LFD) or a high-fat diet (HFD) for 4 weeks, starting at 8 weeks of age. As expected, HFD increased fasting hyperinsulinemia, and Ins1+/- mice had significantly lower circulating insulin compared with Ins1+/+ littermate controls. Fasting glucose and body weight were not significantly different between genotypes at any time over the 4 weeks of study. In liver and skeletal muscle, protein abundances reflecting complex I (Ndufb8), II (Sdhb), III (Uqcrc2), and V (Atp5a1) were not consistently different between groups. In mesenteric white adipose tissue, Ins1+/- mice had reduced abundance of Ndufb8 and Sdhb proteins. Ucp1 protein abundance was increased in the context of the HFD, and HFD alone had a dramatic inhibitory effect on Pparg protein levels. In inguinal white adipose tissue, Ins1+/- mice exhibited significant increases in all oxidative mitochondrial complexes measured, independent of diet. No changes in Ucp1 or Pparg protein, or Prdm16:Pparg association were found. While HFD increased the abundance of nuclear Sirt1, no effects on total Sirt3 protein levels were observed in this tissue. In brown adipose tissue, lowered insulin increased Sdhb protein levels that had been reduced by HFD. Ucp1 protein levels, Prdm16:Pparg association, and Sirt3 abundance were all increased in the absence of diet-induced hyperinsulinemia. Our data show that in young mice, reducing insulin upregulates oxidative proteins in inguinal fat without affecting Ucp1, while in mesenteric white fat and brown adipose tissue, reducing insulin upregulates Ucp1 in the context of HFD. Collectively, our results show that preventing hyperinsulinemia has depot-specific effects on adipose tissue metabolism and helps explain the increased energy expenditure previously reported in Ins1+/- mice.
- Published
- 2020
- Full Text
- View/download PDF
16. Elucidation of the 14-3-3ζ interactome reveals critical roles of RNA-splicing factors during adipogenesis
- Author
-
Mina Sadeghi, Thibault Mayor, Gareth E. Lim, Nancy N. Fang, and Yves Mugabo
- Subjects
Proteomics ,0301 basic medicine ,Scaffold protein ,RNA Splicing Factors ,Mice, Transgenic ,Biology ,Biochemistry ,Interactome ,Mice ,03 medical and health sciences ,PTRF ,Pregnancy ,3T3-L1 Cells ,Protein Interaction Mapping ,Adipocytes ,Animals ,Protein Interaction Maps ,RNA, Messenger ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Cells, Cultured ,Adipogenesis ,Effector ,Alternative splicing ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Embryo, Mammalian ,Cell biology ,PPAR gamma ,Alternative Splicing ,030104 developmental biology ,14-3-3 Proteins ,RNA splicing ,Female ,Signal Transduction - Abstract
Adipogenesis involves a complex signaling network requiring strict temporal and spatial organization of effector molecules. Molecular scaffolds, such as 14-3-3 proteins, facilitate such organization, and we have previously identified 14-3-3ζ as an essential scaffold in adipocyte differentiation. The interactome of 14-3-3ζ is large and diverse, and it is possible that novel adipogenic factors may be present within it, but this possibility has not yet been tested. Herein, we generated mouse embryonic fibroblasts from mice overexpressing a tandem affinity purification (TAP) epitope–tagged 14-3-3ζ molecule. After inducing adipogenesis, TAP–14-3-3ζ complexes were purified, followed by MS analysis to determine the 14-3-3ζ interactome. We observed more than 100 proteins that were unique to adipocyte differentiation, 56 of which were novel interacting partners. Among these, we were able to identify previously established regulators of adipogenesis (i.e. Ptrf/Cavin1) within the 14-3-3ζ interactome, confirming the utility of this approach to detect adipogenic factors. We found that proteins related to RNA metabolism, processing, and splicing were enriched in the interactome. Analysis of transcriptomic data revealed that 14-3-3ζ depletion in 3T3-L1 cells affected alternative splicing of mRNA during adipocyte differentiation. siRNA-mediated depletion of RNA-splicing factors within the 14-3-3ζ interactome, that is, of Hnrpf, Hnrpk, Ddx6, and Sfpq, revealed that they have essential roles in adipogenesis and in the alternative splicing of Pparg and the adipogenesis-associated gene Lpin1. In summary, we have identified novel adipogenic factors within the 14-3-3ζ interactome. Further characterization of additional proteins within the 14-3-3ζ interactome may help identify novel targets to block obesity-associated expansion of adipose tissues.
- Published
- 2018
- Full Text
- View/download PDF
17. 14-3-3ζ mediates an alternative, non-thermogenic mechanism to reduce heat loss and improve cold tolerance
- Author
-
Alain Rivard, Kadidia Diallo, André C. Carpentier, Angel F. Lopez, Gareth E. Lim, Christophe Noll, and Sylvie Dussault
- Subjects
0303 health sciences ,medicine.medical_specialty ,Mechanism (biology) ,Chemistry ,Transgene ,Wild type ,Heat losses ,Endogeny ,Norepinephrine (medication) ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Homeothermy ,Thermogenesis ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
SummaryFollowing prolonged cold exposure, adaptive thermogenic pathways are activated to maintain homeothermy, and elevations in body temperature are generally associated with UCP1-dependent and -independent increases in energy expenditure. One of the earliest, identified functions of the molecular scaffold, 14-3-3ζ, was its role in the synthesis of norepinephrine, a key endogenous factor that stimulates thermogenesis. This suggests that 14-3-3ζ may have critical roles in cold-induced thermogenesis. Herein, we report that transgenic over-expression of TAP-14-3-3ζ in mice significantly improved tolerance to prolonged cold. When compared to wildtype controls, TAP mice displayed significantly elevated body temperatures and paradoxical decreases in energy expenditure. No changes in β-adrenergic sensitivity or oxidative metabolism were observed; instead, 14-3-3ζ over-expression significantly decreased thermal conductance via increased peripheral vasoconstriction. These findings suggest 14-3-3ζ mediates alternative, non-thermogenic mechanisms to mitigate heat loss for homeothermy. Our results point to an unexpected role of 14-3-3ζ in the regulation of body temperature.Graphical abstract
- Published
- 2019
- Full Text
- View/download PDF
18. 2034-P: The Role of 14-3-3zeta in the 'Beiging' of White Adipose Tissue
- Author
-
Kadidia Diallo and Gareth E. Lim
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,White adipose tissue ,Biology ,chemistry.chemical_compound ,Endocrinology ,Lipid oxidation ,chemistry ,Adipogenesis ,Adipocyte ,Internal medicine ,Knockout mouse ,Internal Medicine ,medicine ,Glucose homeostasis ,Adipocyte hypertrophy - Abstract
Adipocyte hypertrophy and hyperplasia is a hallmark of obesity, and treatments that induce the oxidation of lipid stores may represent a potential therapy. When stimulated by cold or beta-adrenergic agonists, adipocytes in inguinal white adipose tissue (iWAT) can convert to beige cells, which resemble brown adipocytes. This process can reduce body weight in rodents and improve glucose homeostasis through increased Ucp1-dependent lipid oxidation. 14-3-3?, a molecular scaffold we found to be essential for adipogenesis, regulates the enzymatic activities of tryptophan and tyrosine hydroxylases, both of which influence beiging. Thus, the aim of the current study is to investigate whether 14-3-3? influences the beiging process. Compared to control mice, acute (3 hr) and chronic (72 hr) cold (4 C) exposure in male transgenic mice over-expressing 14-3-3? led to improved cold tolerance due to significantly increased Ucp1 mRNA and protein in iWAT. Following chronic exposure, they were also able to maintain their body weight by increasing their food intake. Consistent with these data, analysis of adipocyte area revealed a decrease in the size of inguinal adipocytes in transgenic mice, suggesting increased lipid oxidation. In contrast, gonadal adipocytes were larger in transgenic mice, which may explain their ability to maintain their body weight. Systemic 14-3-3? knockout mice had significantly lower levels of Ucp1 mRNA in iWAT and BAT but did not display differences in tolerance to acute cold. Depletion of 14-3-3? by siRNA in brown adipocytes did not impair isoproterenol-mediated induction of Ucp1 mRNA, suggesting that 14-3-3? is not required for Ucp1 expression in this cell type. In future studies, we will examine if 14-3-3? influences the responsiveness of iWAT to chronic β-adrenergic stimuli and assess the impact of 14-3-3? overexpression in brown adipocyte function. Collectively, our results point to a novel role of 14-3-3? in the beiging of inguinal adipocytes and increase our understanding of how beiging is regulated. Disclosure K. Diallo: None. G.E. Lim: None. Funding Canadian Institutes of Health Research; Fonds de la recherche en santé du Québec; Centre de recherche du CHUM; Montreal Diabetes Research Center
- Published
- 2019
- Full Text
- View/download PDF
19. 2015-P: 14-3-3zeta Is Required for PKA-Dependent Lipolysis
- Author
-
Abel Oppong, Gareth E. Lim, and Yves Mugabo
- Subjects
medicine.medical_specialty ,Gene knockdown ,Peroxisome proliferator-activated receptor gamma ,Forskolin ,biology ,Endocrinology, Diabetes and Metabolism ,CREB ,Monoacylglycerol lipase ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Adipogenesis ,Adipocyte ,Internal medicine ,Internal Medicine ,medicine ,biology.protein ,Lipolysis - Abstract
The molecular scaffold, 14-3-3?, was previously found to be essential for visceral adipogenesis, but its contributions to the function of mature adipocytes is not known. As it can regulate the activities of metabolic effectors, we hypothesized that 14-3-3? also has essential roles in adipocyte function. 3T3-L1 adipocytes and mouse models were used to study if 14-3-3? regulates lipolysis. Depletion of 14-3-3? by siRNA abrogated glycerol and free fatty acid (FFA) release from 3T3-L1 cells treated with Isoproterenol (ISO, 1 μM), Forskolin (FSK, 10 μM), and dibutyryl cAMP (1 mM). In contrast, over-expression of 14-3-3? potentiated ISO-mediated FFA release. Knockdown of 14-3-3? did not affect cAMP generation in ISO- and FSK-treated 3T3-L1 cells, but mRNA levels of lipases (Atgl, Hsl, and Magl) and Pparg were reduced, suggesting a loss of adipocyte identity. Decreased activation and total expression of PKA substrates, including Hsl and CREB, were detected in 14-3-3?-depleted 3T3-L1 cells. Taken together, these data suggest that 14-3-3? is necessary for lipolysis from 3T3-L1 adipocytes. To understand adipocyte-specific roles of 14-3-3?, tamoxifen (TMX)-inducible, adipocyte-specific 14-3-3? knockout (adi14-3-3?KO) mice were used. Four weeks after TMX exposure (5 days, 50 mg/kg), no effects on body weight were found. After an overnight fast, adi14-3-3?KO mice displayed impaired lipolysis following i.p CL-316,243 (1 mg/kg) injections. In contrast, transgenic over-expression of 14-3-3? did not affect lipolysis. Adi14-3-3?KO mice also displayed glucose intolerance following i.p. glucose (2 g/kg). Real-time PCR confirmed significant reductions in Atgl, Hsl, and Pparg mRNA levels in adi14-3-3?KO mice, suggesting impaired adipocyte function. Collectively, these results demonstrate essential functions of 14-3-3? in facilitating lipolysis and, potentially, adipocyte maturity. Future studies are aimed at understanding how 14-3-3? regulates other aspects of adipocyte function, including diet-induced expansion of fat mass. Disclosure A. Oppong: None. Y. Mugabo: None. G.E. Lim: None. Funding Canadian Institutes of Health Research
- Published
- 2019
- Full Text
- View/download PDF
20. Scaffold Proteins: From Coordinating Signaling Pathways to Metabolic Regulation
- Author
-
Gareth E. Lim and Yves Mugabo
- Subjects
0301 basic medicine ,Scaffold protein ,Blood Glucose ,medicine.medical_specialty ,Context (language use) ,Apoptosis ,Disease ,Biology ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Metabolic Diseases ,Nuclear Matrix-Associated Proteins ,Internal medicine ,Insulin Secretion ,medicine ,Diabetes Mellitus ,Glucose homeostasis ,Animals ,Homeostasis ,Humans ,Obesity ,Adiposity ,Effector ,Kinase ,Gluconeogenesis ,Mini-Review ,3. Good health ,030104 developmental biology ,Cardiovascular Diseases ,Signal transduction ,Insulin Resistance ,Energy Metabolism ,030217 neurology & neurosurgery ,Function (biology) ,Signal Transduction - Abstract
Among their pleiotropic functions, scaffold proteins are required for the accurate coordination of signaling pathways. It has only been within the past 10 years that their roles in glucose homeostasis and metabolism have emerged. It is well appreciated that changes in the expression or function of signaling effectors, such as receptors or kinases, can influence the development of chronic diseases such as diabetes and obesity. However, little is known regarding whether scaffolds have similar roles in the pathogenesis of metabolic diseases. In general, scaffolds are often underappreciated in the context of metabolism or metabolic diseases. In the present review, we discuss various scaffold proteins and their involvement in signaling pathways related to metabolism and metabolic diseases. The aims of the present review were to highlight the importance of scaffold proteins and to raise awareness of their physiological contributions. A thorough understanding of how scaffolds influence metabolism could aid in the discovery of novel therapeutic approaches to treat chronic conditions, such as diabetes, obesity, and cardiovascular disease, for which the incidence of all continue to increase at alarming rates.
- Published
- 2018
21. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain
- Author
-
Derek A. Dionne, Melissa M. Page, James D. Johnson, Søs Skovsø, Marta Szabat, Stephane Flibotte, Sunita Sinha, Brian Rodrigues, Gareth E. Lim, Haoning Cen, Daria Hutchinson, Amy Pei-Ling Chiu, and Corey Nislow
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,obesity ,medicine.medical_treatment ,Adipose tissue ,Biology ,Diet, High-Fat ,Weight Gain ,Biochemistry ,03 medical and health sciences ,Mice ,PTRF ,Weight loss ,Internal medicine ,Genetics ,medicine ,Hyperinsulinemia ,Glucose homeostasis ,Animals ,Homeostasis ,Insulin ,Molecular Biology ,innate immunity ,Adiposity ,2. Zero hunger ,Mice, Knockout ,Research ,Body Weight ,Ptrf/Cavin ,medicine.disease ,adipose tissue ,030104 developmental biology ,Endocrinology ,high-fat diet ,medicine.symptom ,Lipodystrophy ,Weight gain ,Gene Deletion ,Biotechnology - Abstract
Excess circulating insulin is associated with obesity in humans and in animal models. However, the physiologic causality of hyperinsulinemia in adult obesity has rightfully been questioned because of the absence of clear evidence that weight loss can be induced by acutely reversing diet-induced hyperinsulinemia. Herein, we describe the consequences of inducible, partial insulin gene deletion in a mouse model in which animals have already been made obese by consuming a high-fat diet. A modest reduction in insulin production/secretion was sufficient to cause significant weight loss within 5 wk, with a specific effect on visceral adipose tissue. This result was associated with a reduction in the protein abundance of the lipodystrophy gene polymerase I and transcript release factor ( Ptrf; Cavin) in gonadal adipose tissue. RNAseq analysis showed that reduced insulin and weight loss also associated with a signature of reduced innate immunity. This study demonstrates that changes in circulating insulin that are too fine to adversely affect glucose homeostasis nonetheless exert control over adiposity.-Page, M. M., Skovso, S., Cen, H., Chiu, A. P., Dionne, D. A., Hutchinson, D. F., Lim, G. E., Szabat, M., Flibotte, S., Sinha, S., Nislow, C., Rodrigues, B., Johnson, J. D. Reducing insulin via conditional partial gene ablation in adults reverses diet-induced weight gain.
- Published
- 2017
22. Elucidation of the 14-3-3ζ interactome reveals critical roles of RNA splicing factors during adipogenesis
- Author
-
Nancy N. Fang, Mina Sadeghi, Thibault Mayor, Gareth E. Lim, and Yves Mugabo
- Subjects
RNA Splicing Factors ,Genetics ,0303 health sciences ,Effector ,Alternative splicing ,Biology ,Interactome ,Cell biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,PTRF ,Adipogenesis ,RNA splicing ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Adipogenesis is facilitated by a complex signaling network requiring strict temporal and spatial organization of effector molecules. Molecular scaffolds, such as 14-3-3 proteins, coordinate such events, and we have previously identified 14-3-3ζ as an essential scaffold in adipocyte differentiation. The interactome of 14-3-3ζ is large and diverse, and it is possible that novel adipogenic factors may be present within it. Mouse embryonic fibroblasts from mice over-expressing a TAP-epitope-tagged 14-3-3ζ molecule were generated, and following the induction of adipogenesis, TAP-14-3-3ζ complexes were purified, followed by mass spectrometry analysis to determine the 14-3-3ζ interactome. Over 100 proteins were identified as being unique to adipocyte differentiation, of which 56 were novel interacting partners. Previously established regulators of adipogenesis (ie, Ptrf/Cavin1 and Phb2) were found within the 14-3-3ζ interactome, confirming the ability of this approach to identify regulators of adipocyte differentiation. An enrichment of proteins in the interactome related to RNA metabolism, processing, and splicing was identified, and analysis of transcriptomic data revealed that 14-3-3ζ depletion in 3T3-L1 cells affected the alternative splicing of mRNA during adipocyte differentiation. Of the RNA splicing factors within the 14-3-3ζ interactome, depletion of Hnrnpf, Hnrnpk, Ddx6, and Sfpq by siRNA revealed essential roles of these proteins in adipogenesis and their roles in the alternative splicing of Lpin1. In summary, novel adipogenic factors can be detected within the 14-3-3ζ interactome, and further characterization of additional proteins within the 14-3-3ζ interactome has the potential of identifying novel targets to block the expansion of adipose tissue mass that occurs in obesity.
- Published
- 2017
- Full Text
- View/download PDF
23. Role of 14-3-3Zeta in the 'Beiging' of White Adipocytes
- Author
-
Abel Oppong, Kadidia Diallo, Yves Mugabo, and Gareth E. Lim
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Internal medicine ,Internal Medicine ,medicine ,General Medicine ,White Adipocytes ,business - Published
- 2018
- Full Text
- View/download PDF
24. Ywhaz/14-3-3ζ Deletion Improves Glucose Tolerance Through a GLP-1-Dependent Mechanism
- Author
-
James E. Lulo, Angel F. Lopez, Micah Piske, Gareth E. Lim, James D. Johnson, Hayley S. Ramshaw, Lim, Gareth E, Piske, Micah, Lulo, James E, Ramshaw, Hayley S, Lopez, Angel F, and Johnson, James D
- Subjects
0301 basic medicine ,Blood Glucose ,insulin ,medicine.medical_specialty ,mice ,fasting ,glucose tolerance ,medicine.medical_treatment ,Enteroendocrine Cells ,hormone ,Biology ,Glucagon-Like Peptide-1 Receptor ,Endocrinology & Metabolism ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Glucagon-Like Peptide 1 ,Internal medicine ,Insulin-Secreting Cells ,Glucose Intolerance ,medicine ,glucose homeostasis ,Glucose homeostasis ,Animals ,Homeostasis ,Insulin ,glucose ,Phosphorylation ,tube feeding ,Mice, Knockout ,Glucose tolerance test ,intestines ,medicine.diagnostic_test ,Glucose Tolerance Test ,glucagon-like pepide-1 receptor ,glucagon-like peptide 1 ,030104 developmental biology ,14-3-3 Proteins ,Adipogenesis ,030220 oncology & carcinogenesis ,YWHAZ ,Knockout mouse ,YWHAZ Gene ,Proto-Oncogene Proteins c-akt - Abstract
Multiple signaling pathways mediate the actions of metabolic hormones to control glucose homeostasis, but the proteins that coordinate such networks are poorly understood. We previously identified the molecular scaffold protein, 14-3-3 zeta, as a critical regulator of in vitro beta-cell survival and adipogenesis, but its metabolic roles in glucose homeostasis have not been studied in depth. Herein, we report that Ywhaz gene knockout mice (14-3-3 zeta KO) exhibited elevated fasting insulin levels while maintaining normal beta-cell responsiveness to glucose when compared with wild-type littermate controls. In contrast with our observations after an ip glucose bolus, glucose tolerance was significantly improved in 14-3-3 zeta KO mice after an oral glucose gavage. This improvement in glucose tolerance was associated with significantly elevated fasting glucagon-like peptide-1 (GLP-1) levels. 14-3-3 zeta knockdown in GLUTag L cells elevated GLP-1 synthesis and increased GLP-1 release. Systemic inhibition of the GLP-1 receptor attenuated the improvement in oral glucose tolerance that was seen in 14-3-3 zeta KO mice. When taken together these findings demonstrate novel roles of 14-3-3 zeta in the regulation of glucose homeostasis and suggest that modulating 14-3-3 zeta levels in intestinal L cells may have beneficial metabolic effects through GLP-1-dependent mechanisms. Refereed/Peer-reviewed
- Published
- 2016
25. Hyperinsulinemia Drives Diet-Induced Obesity Independently of Brain Insulin Production
- Author
-
James D. Johnson, Gareth E. Lim, Shernaz X. Bamji, Nicole M. Templeman, Ali Asadi, José Diego Botezelli, Kwan-Yi Chu, G. Stefano Brigidi, Susanne M. Clee, Bradford G. Hoffman, Xiaoke Hu, Arya E. Mehran, and Timothy J. Kieffer
- Subjects
medicine.medical_specialty ,endocrine system ,Pancreatic islet hyperplasia ,endocrine system diseases ,Physiology ,medicine.medical_treatment ,Adipose tissue ,030209 endocrinology & metabolism ,White adipose tissue ,Carbohydrate metabolism ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Adipocyte ,medicine ,Hyperinsulinemia ,Molecular Biology ,030304 developmental biology ,2. Zero hunger ,0303 health sciences ,Insulin ,nutritional and metabolic diseases ,Cell Biology ,medicine.disease ,Thermogenin ,Endocrinology ,chemistry - Abstract
Summary Hyperinsulinemia is associated with obesity and pancreatic islet hyperplasia, but whether insulin causes these phenomena or is a compensatory response has remained unsettled for decades. We examined the role of insulin hypersecretion in diet-induced obesity by varying the pancreas-specific Ins1 gene dosage in mice lacking Ins2 gene expression in the pancreas, thymus, and brain. Age-dependent increases in fasting insulin and β cell mass were absent in Ins1 +/− : Ins2 −/− mice fed a high-fat diet when compared to Ins1 +/+ : Ins2 −/− littermate controls. Remarkably, Ins1 +/− : Ins2 −/− mice were completely protected from diet-induced obesity. Genetic prevention of chronic hyperinsulinemia in this model reprogrammed white adipose tissue to express uncoupling protein 1 and increase energy expenditure. Normalization of adipocyte size and activation of energy expenditure genes in white adipose tissue was associated with reduced inflammation, reduced fatty acid spillover, and reduced hepatic steatosis. Thus, we provide genetic evidence that pathological circulating hyperinsulinemia drives diet-induced obesity and its complications.
- Published
- 2012
- Full Text
- View/download PDF
26. Identification of Novel Adipogenic Factors in the 14–3-3ζ Interactome During Adipocyte Differentiation
- Author
-
Thibault Mayor, Gareth E. Lim, James D. Johnson, Amparo Acker-Palmer, Jason T. C. Lee, Yves Mugabo, and Nancy N. Fang
- Subjects
business.industry ,Endocrinology, Diabetes and Metabolism ,General Medicine ,Computational biology ,Interactome ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Adipogenesis ,Adipocyte ,Internal Medicine ,Medicine ,Identification (biology) ,business - Published
- 2017
- Full Text
- View/download PDF
27. 14-3-3Zeta: A Regulator of Lipolysis?
- Author
-
Mina Sadeghi, Yves Mugabo, Abel Oppong, Kadidia Diallo, and Gareth E. Lim
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Internal medicine ,Internal Medicine ,medicine ,Regulator ,Lipolysis ,General Medicine ,business - Published
- 2018
- Full Text
- View/download PDF
28. The Rho Guanosine 5′-Triphosphatase, Cell Division Cycle 42, Is Required for Insulin-Induced Actin Remodeling and Glucagon-Like Peptide-1 Secretion in the Intestinal Endocrine L Cell
- Author
-
Patricia L. Brubaker, Jane Sun, Molie Xu, Tianru Jin, and Gareth E. Lim
- Subjects
MAPK/ERK pathway ,medicine.medical_specialty ,Cell division ,Insulin ,medicine.medical_treatment ,Actin remodeling ,Enteroendocrine cell ,RAC1 ,macromolecular substances ,CDC42 ,Biology ,Endocrinology ,Internal medicine ,medicine ,Secretion - Abstract
Rho GTPases, such as cell division cycle 42 (Cdc42) and ras-related C3 botulinum toxin substrate 1 (Rac1), have been identified as regulators of F-actin dynamics and hormone release from endocrine cells; however, their role in secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1), from the enteroendocrine L cell is unknown. Insulin induced a 1.4-fold increase in L cell GLP-1 release; however, secretion was potentiated to 2.1-fold in the presence of the F-actin depolymerizing agent, latrunculin B, suggesting that F-actin functions as a permissive barrier. In murine GLUTag L cells, insulin stimulated F-actin depolymerization and Cdc42 activation simultaneously, and these events occurred prior to detectable increases in insulin-induced GLP-1 release. After insulin treatment, Cdc42-dependent p21-activated kinase-1 (PAK1) activation was also detected, and transfection of small-interfering RNA against Cdc42 or of dominant-negative Cdc42(T17N) impaired insulin-stimulated PAK1 activation, actin remodeling, and GLP-1 secretion. Overexpression of kinase-dead PAK1(K299R) or PAK1 small interfering RNA similarly attenuated insulin-induced GLP-1 secretion. Knockdown or inhibition of Cdc42 and PAK1 activities also prevented activation of MAPK/ERK (MEK)-1/2-ERK1/2 by insulin, which was previously identified as a critical pathway for insulin-regulated GLP-1 release. Taken together, these data identify a novel signaling pathway in the endocrine L cell, whereby Cdc42 regulates actin remodeling, activation of the cannonical 1/2-ERK1/2 pathway and PAK1, and GLP-1 secretion in response to insulin.
- Published
- 2009
- Full Text
- View/download PDF
29. Insulin Regulates Glucagon-Like Peptide-1 Secretion from the Enteroendocrine L Cell
- Author
-
Christopher J. Rhodes, Nina Flora, Gareth E. Lim, Patricia L. Brubaker, Derek LeRoith, and Guan J. Huang
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Enteroendocrine Cells ,medicine.medical_treatment ,Receptor, IGF Type 1 ,Mice ,Endocrinology ,Insulin resistance ,Glucagon-Like Peptide 1 ,Pregnancy ,Internal medicine ,Insulin receptor substrate ,Diabetes - Insulin - Glucagon - Gastrointestinal ,medicine ,Animals ,Humans ,Insulin ,Insulin-Like Growth Factor I ,Rats, Wistar ,Protein kinase B ,Cells, Cultured ,Mice, Knockout ,biology ,medicine.disease ,Glucagon-like peptide-1 ,Receptor, Insulin ,IRS2 ,Rats ,Insulin oscillation ,Insulin receptor ,biology.protein ,Female ,Insulin Resistance ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Insulin resistance and type 2 diabetes mellitus are associated with impaired postprandial secretion of glucagon-like peptide-1 (GLP-1), a potent insulinotropic hormone. The direct effects of insulin and insulin resistance on the L cell are unknown. We therefore hypothesized that the L cell is responsive to insulin and that insulin resistance impairs GLP-1 secretion. The effects of insulin and insulin resistance were examined in well-characterized L cell models: murine GLUTag, human NCI-H716, and fetal rat intestinal cells. MKR mice, a model of chronic hyperinsulinemia, were used to assess the function of the L cell in vivo. In all cells, insulin activated the phosphatidylinositol 3 kinase-Akt and MAPK kinase (MEK)-ERK1/2 pathways and stimulated GLP-1 secretion by up to 275 ± 58%. Insulin resistance was induced by 24 h pretreatment with 10−7m insulin, causing a marked reduction in activation of Akt and ERK1/2. Furthermore, both insulin-induced GLP-1 release and secretion in response to glucose-dependent insulinotropic peptide and phorbol-12-myristate-13-acetate were significantly attenuated. Whereas inhibition of phosphatidylinositol 3 kinase with LY294002 potentiated insulin-induced GLP-1 release, secretion was abrogated by inhibiting the MEK-ERK1/2 pathway with PD98059 or by overexpression of a kinase-dead MEK1-ERK2 fusion protein. Compared with controls, MKR mice were insulin resistant and displayed significantly higher fasting plasma insulin levels. Furthermore, they had significantly higher basal GLP-1 levels but displayed impaired GLP-1 secretion after an oral glucose challenge. These findings indicate that the intestinal L cell is responsive to insulin and that insulin resistance in vitro and in vivo is associated with impaired GLP-1 secretion. Insulin is a novel secretagogue of the incretin hormone, glucagon-like peptide-1 (GLP-1), and L cell insulin resistance impairs heterologous secretagogue-induced GLP-1 secretion in vitro and in vivo.
- Published
- 2009
- Full Text
- View/download PDF
30. 14-3-3ζ: A numbers game in adipocyte function?
- Author
-
Gareth E. Lim and James D. Johnson
- Subjects
0301 basic medicine ,Cell signaling ,insulin ,Histology ,Protein family ,Fourteen-3-3 ,Regulator ,Context (language use) ,Biology ,scaffold ,adipocyte ,adipogenesis ,14-3-3ζ ,03 medical and health sciences ,0302 clinical medicine ,Commentaries ,Glucose homeostasis ,Transcription factor ,browning ,Cell Biology ,differentiation ,Cell biology ,030104 developmental biology ,Biochemistry ,Adipogenesis ,lipolysis ,Signal transduction ,GLUT4 ,030217 neurology & neurosurgery - Abstract
Molecular scaffolds are often viewed as passive signaling molecules that facilitate protein-protein interactions. However, new evidence gained from the use of loss-of-function or gain-of-function models is dispelling this notion. Our own recent discovery of 14-3-3ζ as an essential regulator of adipogenesis highlights the complex roles of this member of the 14-3-3 protein family. Depletion of the 14-3-3ζ isoform affected parallel pathways that drive adipocyte development, including pathways controlling the stability of key adipogenic transcription factors and cell cycle progression. Going beyond adipocyte differentiation, this study opens new avenues of research in the context of metabolism, as 14-3-3ζ binds to a variety of well-established metabolic proteins that harbor its canonical phosphorylation binding motifs. This suggests that 14-3-3ζ may contribute to key metabolic signaling pathways, such as those that facilitate glucose uptake and fatty acid metabolism. Herein, we discuss these novel areas of research, which will undoubtedly shed light onto novel roles of 14-3-3ζ, and perhaps its related family members, on glucose homeostasis.
- Published
- 2015
31. Glucagon-Like Peptide 1 Secretion by the L-Cell
- Author
-
Gareth E. Lim and Patricia L. Brubaker
- Subjects
endocrine system ,medicine.medical_specialty ,Gastric emptying ,Endocrinology, Diabetes and Metabolism ,Leptin ,digestive, oral, and skin physiology ,Biology ,Glucagon ,Glucagon-like peptide-1 ,Energy homeostasis ,Endocrinology ,Gastrointestinal hormone ,Internal medicine ,Internal Medicine ,medicine ,Secretion ,hormones, hormone substitutes, and hormone antagonists ,Hormone - Abstract
Glucagon-like peptide 1 (GLP-1) is a gut-derived peptide secreted from intestinal L-cells after a meal. GLP-1 has numerous physiological actions, including potentiation of glucose-stimulated insulin secretion, enhancement of β-cell growth and survival, and inhibition of glucagon release, gastric emptying, and food intake. These antidiabetic effects of GLP-1 have led to intense interest in the use of this peptide for the treatment of patients with type 2 diabetes. Oral nutrients such as glucose and fat are potent physiological regulators of GLP-1 secretion, but non-nutrient stimulators of GLP-1 release have also been identified, including the neuromodulators acetylcholine and gastrin-releasing peptide. Peripheral hormones that participate in energy homeostasis, such as leptin, have also been implicated in the regulation of GLP-1 release. Recent studies have begun to elucidate the intracellular signaling pathways that mediate the effects of GLP-1 secretagogues on the intestinal L-cell. The purpose of this review is to summarize the known signaling mechanisms of GLP-1 secretagogues based on the available literature. A better understanding of the pathways underlying GLP-1 secretion may lead to novel approaches by which the levels of this important insulinotropic hormone can be enhanced in patients with type 2 diabetes.
- Published
- 2006
- Full Text
- View/download PDF
32. Fetal and neonatal exposure to nicotine in Wistar rats results in increased beta cell apoptosis at birth and postnatal endocrine and metabolic changes associated with type 2 diabetes
- Author
-
Hertzel C. Gerstein, Jim Petrik, Alison C. Holloway, Gareth E. Lim, Warren G. Foster, and Katherine M. Morrison
- Subjects
Nicotine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Apoptosis ,Type 2 diabetes ,Biology ,Random Allocation ,Fetus ,Pregnancy ,Insulin-Secreting Cells ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Insulin ,Endocrine system ,Glucose homeostasis ,Obesity ,Rats, Wistar ,Adiposity ,Hemostasis ,Glucose tolerance test ,medicine.diagnostic_test ,Body Weight ,Glucose Tolerance Test ,medicine.disease ,Rats ,Disease Models, Animal ,Glucose ,Endocrinology ,Animals, Newborn ,Diabetes Mellitus, Type 2 ,Prenatal Exposure Delayed Effects ,Pregnancy, Animal ,Female ,medicine.drug - Abstract
Epidemiological studies report an increased risk of obesity and type 2 diabetes in children born to women who smoked during pregnancy. This study examines the effect of fetal and neonatal exposure to nicotine, the major addictive component of cigarettes, on postnatal growth, adiposity and glucose homeostasis.Female Wistar rats were given either saline (vehicle) or nicotine (1 mg kg(-1) day(-1)) during pregnancy and lactation. Serum and pancreas tissue were collected from the infant rats at birth. Postnatal growth was assessed weekly until the rats reached 26 weeks of age and glucose homeostasis was examined by OGTTs performed at 7 and 26 weeks of age.Exposure to nicotine resulted in increased postnatal growth and adiposity. Nicotine exposure also resulted in dysglycaemia at 7 and 26 weeks of age. Serum insulin concentrations were decreased in the pups exposed to nicotine at birth. This was associated with increased beta cell apoptosis (pups of saline-treated mothers 8.8+/-1.21% apoptotic beta cells; pups of nicotine-treated mothers 27.8+/-3.1% apoptotic beta cells).Fetal and neonatal exposure to nicotine results in metabolic changes in the offspring that are consistent with obesity and type 2 diabetes. We propose that these metabolic changes may be a consequence of the initial insult to the beta cell during fetal life and that this animal model has many characteristics of diabetes in humans.
- Published
- 2005
- Full Text
- View/download PDF
33. The Effects of In Utero and Lactational Exposure to Chloroform on Postnatal Growth and Glucose Tolerance in Male Wistar Rats
- Author
-
Sandra I. Stals, Jim Petrik, Alison C. Holloway, Warren G. Foster, and Gareth E. Lim
- Subjects
medicine.medical_specialty ,Fetus ,Glucose tolerance test ,medicine.diagnostic_test ,Offspring ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Biology ,medicine.disease ,Endocrinology ,In utero ,Internal medicine ,Diabetes mellitus ,medicine ,Glucose homeostasis ,Weaning - Abstract
Water chlorination results in the formation of trihalomethanes (THMs) including chloroform. In human studies, fetal growth restriction has been associated with exposure to THMs during pregnancy and impaired fetal growth has been associated with an increased risk of type 2 diabetes. Therefore, the objective of this study was to determine the effect of in utero and lactational exposure to chloroform on birthweight and postnatal indicators of type 2 diabetes. Female Wistar rats were given chloroform (0 µg/L, 75 µg/L) in their drinking water for 2 wk prior to mating until parturition (in utero exposure only) or until weaning (in utero + lactational exposure). At postnatal d 1 (PND1) pups of dams exposed to chloroform had significantly higher serum glucose levels and lower insulin levels, but this effect was not due to β-cell depletion in the neonatal pancreas. Glucose homeostasis in response to a glucose challenge was not changed by chloroform treatment. Chloroform exposure did not affect birthweight; however, offspring of dams exposed to chloroform had significantly impaired postnatal growth. Although fetal and neonatal exposure to chloroform did not elicit physiological changes associated with the onset of type 2 diabetes, there were physiological changes resulting in impaired postnatal growth.
- Published
- 2004
- Full Text
- View/download PDF
34. 14-3-3ζ coordinates adipogenesis of visceral fat
- Author
-
Mark A. Guthridge, Hayley S. Ramshaw, Amparo Acker-Palmer, Gareth E. Lim, Micah Piske, Tobias Albrecht, Susanne M. Clee, Sunita Sinha, James D. Johnson, Corey Nislow, Jason T. C. Lee, Angel F. Lopez, Karnjit Sarai, Lim, Gareth E, Albrecht, Tobias, Piske, Micah, Sarai, Karnjit, Lee, Jason TC, Ramshaw, Hayley S, Sinha, Sunita, Guthridge, Mark A, Acker-Palmer, Amparo, Lopez, Angel F, Clee, Susanne M, Nislow, Corey, and Johnson, James D
- Subjects
CCAAT-Enhancer-Binding Protein-delta ,General Physics and Astronomy ,Peroxisome proliferator-activated receptor ,medical research ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Adipocyte ,Mice, Knockout ,chemistry.chemical_classification ,0303 health sciences ,Gene knockdown ,Adipogenesis ,Multidisciplinary ,Ccaat-enhancer-binding proteins ,biological sciences ,Signal transducing adaptor protein ,Flow Cytometry ,Cell biology ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction ,Immunoblotting ,Kruppel-Like Transcription Factors ,Nerve Tissue Proteins ,In Vitro Techniques ,Intra-Abdominal Fat ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Downregulation and upregulation ,Zinc Finger Protein Gli3 ,3T3-L1 Cells ,Autophagy ,Animals ,Hedgehog Proteins ,Obesity ,Transcription factor ,030304 developmental biology ,Gene Expression Profiling ,General Chemistry ,Molecular biology ,PPAR gamma ,14-3-3 Proteins ,Microscopy, Fluorescence ,chemistry ,CCAAT-Enhancer-Binding Proteins ,NIH 3T3 Cells ,030217 neurology & neurosurgery - Abstract
The proteins that coordinate complex adipogenic transcriptional networks are poorly understood. 14-3-3ζ is a molecular adaptor protein that regulates insulin signalling and transcription factor networks. Here we report that 14-3-3ζ-knockout mice are strikingly lean from birth with specific reductions in visceral fat depots. Conversely, transgenic 14-3-3ζ overexpression potentiates obesity, without exacerbating metabolic complications. Only the 14-3-3ζ isoform is essential for adipogenesis based on isoform-specific RNAi. Mechanistic studies show that 14-3-3ζ depletion promotes autophagy-dependent degradation of C/EBP-δ, preventing induction of the master adipogenic factors, Pparγ and C/EBP-α. Transcriptomic data indicate that 14-3-3ζ acts upstream of hedgehog signalling-dependent upregulation of Cdkn1b/p27Kip1. Indeed, concomitant knockdown of p27Kip1 or Gli3 rescues the early block in adipogenesis induced by 14-3-3ζ knockdown in vitro. Adipocyte precursors in 14-3-3ζKO embryos also appear to have greater Gli3 and p27Kip1 abundance. Together, our in vivo and in vitro findings demonstrate that 14-3-3ζ is a critical upstream driver of adipogenesis., 14-3-3 family proteins are adaptor proteins involved in various cellular functions. Here Lim et al. show that 14-3-3ζ regulates adipogenesis in vitro, and the formation of visceral fat in mice, by reducing autophagic degradation of the adipogenic master transcription factor C/EBP-δ.
- Published
- 2015
35. Effects of insulin on human pancreatic cancer progression modeled in vitro
- Author
-
Yu Hsuan Carol Yang, Corinne A. Hoesli, Michelle Chan, Gareth E. Lim, James D. Johnson, James M. Piret, Emilyn U. Alejandro, Tobias Albrecht, Søs Skovsø, and Garth L. Warnock
- Subjects
MAPK/ERK pathway ,Cancer Research ,Benzylamines ,Indoles ,medicine.medical_treatment ,Pancreatic ductal adenocarcinoma ,Hyperinsulinemia ,PANC1 ,Medicine ,Insulin ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Diabetes ,ERK ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Oncology ,Disease Progression ,Stem cell ,Carcinoma, Pancreatic Ductal ,Research Article ,Cell type ,medicine.medical_specialty ,Cell Survival ,MAP Kinase Signaling System ,Models, Biological ,Phenols ,Internal medicine ,Pancreatic cancer ,Cell Line, Tumor ,Quinoxalines ,Genetics ,Humans ,Protein kinase B ,Cell Proliferation ,Pancreatic duct ,HPDE ,Cell growth ,business.industry ,AKT ,Pancreatic Ducts ,PDAC ,medicine.disease ,Pancreatic Neoplasms ,Proto-Oncogene Proteins c-raf ,Endocrinology ,business ,Proto-Oncogene Proteins c-akt - Abstract
Background: Pancreatic adenocarcinoma is one of the most lethal cancers, yet it remains understudied and poorly understood. Hyperinsulinemia has been reported to be a risk factor of pancreatic cancer, and the rapid rise of hyperinsulinemia associated with obesity and type 2 diabetes foreshadows a rise in cancer incidence. However, the actions of insulin at the various stages of pancreatic cancer progression remain poorly defined. Methods: Here, we examined the effects of a range of insulin doses on signalling, proliferation and survival in three human cell models meant to represent three stages in pancreatic cancer progression: primary pancreatic duct cells, the HPDE immortalized pancreatic ductal cell line, and the PANC1 metastatic pancreatic cancer cell line. Cells were treated with a range of insulin doses, and their proliferation/viability were tracked via live cell imaging and XTT assays. Signal transduction was assessed through the AKT and ERK signalling pathways via immunoblotting. Inhibitors of AKT and ERK signalling were used to determine the relative contribution of these pathways to the survival of each cell model. Results: While all three cell types responded to insulin, as indicated by phosphorylation of AKT and ERK, we found that there were stark differences in insulin-dependent proliferation, cell viability and cell survival among the cell types. High concentrations of insulin increased PANC1 and HPDE cell number, but did not alter primary duct cell proliferation in vitro. Cell survival was enhanced by insulin in both primary duct cells and HPDE cells. Moreover, we found that primary cells were more dependent on AKT signalling, while HPDE cells and PANC1 cells were more dependent on RAF/ERK signalling. Conclusions: Our data suggest that excessive insulin signalling may contribute to proliferation and survival in human immortalized pancreatic ductal cells and metastatic pancreatic cancer cells, but not in normal adult human pancreatic ductal cells. These data suggest that signalling pathways involved in cell survival may be rewired during pancreatic cancer progression.
- Published
- 2014
36. Pancreatic β-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription
- Author
-
Farnaz Taghizadeh, James D. Johnson, Xiaoke Hu, Arya E. Mehran, Dmytro Pelipeychenko, Emilyn U. Alejandro, Gareth E. Lim, and Manuela Baccarini
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,030209 endocrinology & metabolism ,FOXO1 ,Apoptosis ,Biology ,Biochemistry ,Research Communications ,Impaired glucose tolerance ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,Insulin-Secreting Cells ,Insulin Secretion ,Genetics ,medicine ,Glucose homeostasis ,Animals ,Homeostasis ,Insulin ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Mice, Knockout ,0303 health sciences ,Forkhead Box Protein O1 ,Forkhead Transcription Factors ,medicine.disease ,3. Good health ,Insulin oscillation ,Proto-Oncogene Proteins c-raf ,Insulin receptor ,Endocrinology ,biology.protein ,Female ,Biotechnology - Abstract
Regulation of glucose homeostasis by insulin depends on pancreatic β-cell growth, survival, and function. Raf-1 kinase is a major downstream target of several growth factors that promote proliferation and survival of many cell types, including the pancreatic β cells. We have previously reported that insulin protects β cells from apoptosis and promotes proliferation by activating Raf-1 signaling in cultured human islets, mouse islets, and MIN6 cells. As Raf-1 activity is critical for basal apoptosis and insulin secretion in vitro, we hypothesized that Raf-1 may play an important role in glucose homeostasis in vivo. To test this hypothesis, we utilized the Cre-loxP recombination system to obtain a pancreatic β-cell-specific ablation of Raf-1 kinase gene (RIPCre+/+:Raf-1flox/flox) and a complete set of littermate controls (RIPCre+/+:Raf-1wt/wt). RIPCre+/+:Raf-1flox/flox mice were viable, and no effects on weight gain were observed. RIPCre+/+:Raf-1flox/flox mice had increased fasting blood glucose levels and impaired glucose tolerance but normal insulin tolerance compared to littermate controls. Insulin secretion in vivo and in isolated islets was markedly impaired, but there was no apparent effect on the exocytosis machinery. However, islet insulin protein and insulin 2 mRNA, but not insulin 1 mRNA, were dramatically reduced in Raf-1-knockout mice. Analysis of insulin 2 knockout mice demonstrated that this reduction in mRNA was sufficient to impair in vivo insulin secretion. Our data further indicate that Raf-1 specifically and acutely regulates insulin 2 mRNA via negative action on Foxo1, which has been shown to selectively control the insulin 2 gene. This work provides the first direct evidence that Raf-1 signaling is essential for the regulation of basal insulin transcription and the supply of releasable insulin in vivo.—Alejandro, E. U., Lim, G. E., Mehran, A. E., Hu, X., Taghizadeh, F., Pelipeychenko, D., Baccarini, M., Johnson, J. D. Pancreatic β-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription.
- Published
- 2011
37. Interactions of grapefruit juice and cardiovascular medications: A potential risk of toxicity
- Author
-
Gareth E, Lim, Timao, Li, and Harpal S, Buttar
- Subjects
Clinical Review - Abstract
Recently, drug interactions with grapefruit juice (GFJ) have received considerable attention from basic scientists, physicians, industry and drug regulatory agencies. GFJ has been shown to inhibit cytochrome P-450 3A4 isoenzyme and P-glycoprotein transporters in the intestine and liver. The GFJ-induced inhibitory effects are considered to be responsible for alterations in drug bioavailability, and pharmacokinetic and pharmacodynamic changes when drugs are ingested concurrently with GFJ. However, little or no interaction is observed when GFJ is taken concomitantly with parentally administered drugs. It is well known that risk factors for cardiovascular disease increase with advancing age, while hepatic metabolic activity decreases in elderly individuals. It is, therefore, possible that the combination of GFJ and cardiovascular medications may pose a health risk, especially in elderly patients. A number of studies have shown interactions of GFJ with cardiovascular drugs such as calcium-channel blockers, angiotensin II receptor antagonists, beta-blockers, and statins. Such interactions are likely to change the pharmacokinetics and pharmacodynamics of these drugs, consequently causing undesirable health effects. Therefore, health care professionals and the public need to be advised of the potential risks associated with the concomitant use of GFJ and interacting medications, especially cardiovascular drugs and agents with a narrow therapeutic index. This review focuses on the adverse interactions of GFJ and cardiovascular medications, and the proposed underlying mechanisms of such interactions.
- Published
- 2009
38. Prenatal exposure to nicotine causes postnatal obesity and altered perivascular adipose tissue function
- Author
-
Yu-Jing Gao, Robert M.K.W. Lee, Zhao-Hua Zeng, Warren G. Foster, Jim Petrik, Gareth E. Lim, and Alison C. Holloway
- Subjects
Male ,medicine.medical_specialty ,Aging ,Nicotine ,Offspring ,Endocrinology, Diabetes and Metabolism ,Medicine (miscellaneous) ,Adipose tissue ,Aorta, Thoracic ,Weight Gain ,Fat pad ,Muscle, Smooth, Vascular ,Endocrinology ,Pregnancy ,medicine.artery ,Internal medicine ,medicine ,Thoracic aorta ,Animals ,Obesity ,Rats, Wistar ,Mesenteric arteries ,Aorta ,Fetus ,business.industry ,Public Health, Environmental and Occupational Health ,Organ Size ,Mesenteric Arteries ,Rats ,medicine.anatomical_structure ,Adipose Tissue ,Prenatal Exposure Delayed Effects ,Hypertension ,Blood Vessels ,Female ,business ,Adrenergic alpha-Agonists ,Food Science ,medicine.drug ,Muscle Contraction - Abstract
GAO, YU-JING, ALISON C. HOLLOWAY, ZHAO-HUA ZENG, GARETH E. LIM, JAMES J. PETRIK, WARREN G. FOSTER, AND ROBERT M.K.W. LEE. Prenatal exposure to nicotine causes postnatal obesity and altered perivascular adipose tissue function. Obes Res. 2005;13: 687–692. Objective: Recent epidemiological studies have shown that there is an increased risk of obesity and hypertension in children born to women who smoked during pregnancy. The aim of this study was to examine the effect of fetal and neonatal exposure to nicotine, the major addictive component of cigarette smoke, on postnatal adiposity and blood vessel function. Research Methods and Procedures: Female Wistar rats were given nicotine or saline (vehicle) during pregnancy and lactation. Postnatal growth was determined in the male offspring from weaning until 26 weeks of age. At 26 weeks of age, fat pad weight and the function of the perivascular adipose tissue (PVAT) in the thoracic aorta and mesenteric arteries were examined. Results: Exposure to nicotine resulted in increased postnatal body weight and fat pad weight and an increased amount of PVAT in the offspring. Contraction of the aorta induced by phenylephrine was significantly attenuated in the presence of PVAT, whereas this effect was not observed in the aortic rings from the offspring of nicotine-exposed dams. Phenylephrine-induced contraction without PVAT was not different between saline- and nicotine-exposed rats. Transfer of solution incubated with PVAT-intact aorta to PVAT-free aorta induced a marked relaxation response in the rats from saline-exposed dams, but this relaxation response was significantly impaired in the rats from nicotine-exposed dams. Discussion: Our results showed that prenatal nicotine exposure increased adiposity and caused an alteration in the modulatory function of PVAT on vascular relaxation response, thus providing insight into the mechanisms underlying the increased prevalence of obesity and hypertension in children exposed to cigarette smoke in utero.
- Published
- 2005
39. The effects of in utero and lactational exposure to chloroform on postnatal growth and glucose tolerance in male Wistar rats
- Author
-
Gareth E, Lim, Sandra I, Stals, James J, Petrik, Warren G, Foster, and Alison C, Holloway
- Subjects
Blood Glucose ,Male ,Analysis of Variance ,Administration, Oral ,Glucose Tolerance Test ,Animals, Suckling ,Rats ,Islets of Langerhans ,Milk ,Animals, Newborn ,Maternal Exposure ,Pregnancy ,Prenatal Exposure Delayed Effects ,Solvents ,Animals ,Female ,Chloroform ,Rats, Wistar ,Water Pollutants, Chemical - Abstract
Water chlorination results in the formation of trihalomethanes (THMs) including chloroform. In human stud-ies, fetal growth restriction has been associated with exposure to THMs during pregnancy and impaired fetal growth has been associated with an increased risk of type 2 diabetes. Therefore, the objective of this study was to determine the effect of in utero and lactational exposure to chloroform on birthweight and postnatal indicators of type 2 diabetes. Female Wistar rats were given chloroform (0 microg/L, 75 microg/L) in their drinking water for 2 wk prior to mating until parturition (in utero exposure only) or until weaning (in utero+lactational exposure). At postnatal d 1 (PND1) pups of dams exposed to chloroform had significantly higher serum glucose levels and lower insulin levels, but this effect was not due to bFONT "Optima"-cell depletion in the neonatal pancreas. Glucose homeostasis in response to a glucose challenge was not changed by chloroform treatment. Chloroform exposure did not affect birthweight; however, offspring of dams exposed to chloroform had significantly impaired postnatal growth. Although fetal and neonatal exposure to chloroform did not elicit physiological changes associated with the onset of type 2 diabetes, there were physiological changes resulting in impaired postnatal growth.
- Published
- 2004
40. 14-3-3 Zeta is Required for Insulin Sensitivity and Glucose Homeostasis In Vivo
- Author
-
Gareth E. Lim, Micah Piske, Hayley S. Ramshaw, Angel F. Lopez, Mark A. Guthridge, and James D. Johnson
- Subjects
medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Immature cells ,Insulin sensitivity ,General Medicine ,medicine.disease ,Endocrinology ,medicine.anatomical_structure ,In vivo ,14-3-3-ZETA ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Glucose homeostasis ,Pancreas ,business - Abstract
s / Can J Diabetes 36 (2012) S2eS22 S18 plasticity of adult pancreatic b-cells. Efforts to harness these immature cells and manipulate their maturation might be one avenue for replacing or repopulating lost/nonfunctional b-cells in the diabetic pancreas.
- Published
- 2012
- Full Text
- View/download PDF
41. 14-3-3zeta Regulates Adipogenesis, Glucose Tolerance and Insulin Sensitivity in Mice
- Author
-
Hayley S. Ramshaw, Susanne M. Clee, Gareth E. Lim, James D. Johnson, Angel F. Lopez, and Mark A. Guthridge
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Adipogenesis ,Endocrinology, Diabetes and Metabolism ,Internal medicine ,Internal Medicine ,Medicine ,Insulin sensitivity ,General Medicine ,business - Published
- 2013
- Full Text
- View/download PDF
42. Caveolin-1 Mediates the Internalization of Functional Insulin Receptors in Beta-Cells
- Author
-
Ivan R. Nabi, James D. Johnson, Gareth E. Lim, and Tobias Albrecht
- Subjects
biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,media_common.quotation_subject ,General Medicine ,Cell biology ,Insulin receptor ,Endocrinology ,Insulin receptor substrate ,Caveolin 1 ,Internal Medicine ,biology.protein ,Medicine ,business ,Internalization ,Beta (finance) ,media_common - Published
- 2012
- Full Text
- View/download PDF
43. Musashi expression in β-cells coordinates insulin expression, apoptosis and proliferation in response to endoplasmic reticulum stress in diabetes
- Author
-
K Y Chu, Yu Hsuan Carol Yang, Z Ao, James D. Johnson, M Szabat, James M. Piret, T B Kalynyak, T J Kieffer, G L Warnock, Gareth E. Lim, Blair K. Gage, and Ali Asadi
- Subjects
Cancer Research ,proliferation ,Cellular differentiation ,Immunology ,Notch signaling pathway ,Apoptosis ,Nerve Tissue Proteins ,030209 endocrinology & metabolism ,Biology ,insulin gene transcription ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Insulin-Secreting Cells ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Insulin ,HES1 ,Cells, Cultured ,Embryonic Stem Cells ,Cell Proliferation ,030304 developmental biology ,Homeodomain Proteins ,Regulation of gene expression ,0303 health sciences ,Gene knockdown ,Receptors, Notch ,Endoplasmic reticulum ,RNA-Binding Proteins ,Cell Differentiation ,differentiation ,Cell Biology ,Endoplasmic Reticulum Stress ,Activins ,Cell biology ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,NUMB ,Unfolded protein response ,Transcription Factor HES-1 ,Original Article ,type 2 diabetes ,ER stress - Abstract
Diabetes is associated with the death and dysfunction of insulin-producing pancreatic β-cells. In other systems, Musashi genes regulate cell fate via Notch signaling, which we recently showed regulates β-cell survival. Here we show for the first time that human and mouse adult islet cells express mRNA and protein of both Musashi isoforms, as well Numb/Notch/Hes/neurogenin-3 pathway components. Musashi expression was observed in insulin/glucagon double-positive cells during human fetal development and increased during directed differentiation of human embryonic stem cells (hESCs) to the pancreatic lineage. De-differentiation of β-cells with activin A increased Msi1 expression. Endoplasmic reticulum (ER) stress increased Msi2 and Hes1, while it decreased Ins1 and Ins2 expression, revealing a molecular link between ER stress and β-cell dedifferentiation in type 2 diabetes. These effects were independent of changes in Numb protein levels and Notch activation. Overexpression of MSI1 was sufficient to increase Hes1, stimulate proliferation, inhibit apoptosis and reduce insulin expression, whereas Msi1 knockdown had the converse effects on proliferation and insulin expression. Overexpression of MSI2 resulted in a decrease in MSI1 expression. Taken together, these results demonstrate overlapping, but distinct roles for Musashi-1 and Musashi-2 in the control of insulin expression and β-cell proliferation. Our data also suggest that Musashi is a novel link between ER stress and the compensatory β-cell proliferation and the loss of β-cell gene expression seen in specific phases of the progression to type 2 diabetes.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.