Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paul Caton is active.

Publication


Featured researches published by Paul Caton.


Journal of Endocrinology | 2010

PPAR control: it's SIRTainly as easy as PGC

Mary C. Sugden; Paul Caton; Mark J. Holness

This review describes recent advances in our knowledge of the regulatory interactions influencing the expression of peroxisome proliferator-activated receptor (PPAR)-regulated genes. We address recent advances highlighting the role of PPARgamma (PPARG) coactivator-1 (PGC-1) and lipin-1 in co-ordinating the expression of genes controlling nutrient handling. We evaluate the possibility that SIRT1 lies at the heart of a regulatory loop involving PPARalpha, PGC-1alpha (PPARA, PPARGC1A as given in the HUGO Database), and lipin-1 (LPIN1 as listed in the HUGO Database) that ultimately controls the metabolic response to varying nutrient and physiological signals via a common mechanism mediated by post-translation modifications (deacetylation) of both PPARalpha and PGC-1s. Finally, we comment on the potential of pharmaceutical manipulation of these targets as well as the possible problems associated with this strategy.


Journal of Endocrinology | 2010

Metformin suppresses hepatic gluconeogenesis through induction of SIRT1 and GCN5

Paul Caton; Nanda Nayuni; Julius Kieswich; Noorafza Q. Khan; Muhammed M. Yaqoob; Roger Corder

Abnormal elevation of hepatic gluconeogenesis is central to the onset of hyperglycaemia in patients with type 2 diabetes mellitus (T2DM). Metformin corrects hyperglycaemia through inhibition of gluconeogenesis, but its mechanism of action is yet to be fully described. SIRT1 and GCN5 (listed as KAT2A in the MGI Database) have recently been identified as regulators of gluconeogenic gene expression through modulation of levels and activity of the coactivators cAMP-response element binding protein-regulated transcription coactivator 2 (TORC2 or CRTC2 as listed in the MGI Database) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC1alpha or PPARGC1A as listed in the MGI Database). We report that in db/db mice, metformin (250 mg/kg per day; 7 days) increases hepatic levels of GCN5 protein and mRNA compared with the untreated db/db mice, as well as increases levels of SIRT1 protein and activity relative to controls and untreated db/db mice. These changes were associated with reduced TORC2 protein level and decreased gene expression and activation of the PGC1alpha gene target phosphoenolpyruvate carboxykinase, and lower plasma glucose and insulin. Inhibition of SIRT1 partially blocked the effects of metformin on gluconeogenesis. SIRT1 was increased through an AMP-activated protein kinase-mediated increase in gene expression of nicotinamide phosphoribosyltransferase, the rate-limiting enzyme of the salvage pathway for NAD(+). Moreover, levels of GCN5 were dramatically reduced in db/db mice compared with the controls. This indicates that loss of GCN5-mediated inhibition of gluconeogenesis appears to constitute a major mechanism for the onset of abnormally elevated hepatic glucose production in db/db mice. In conclusion, induction of GCN5 and SIRT1 potentially represents a critical mechanism of action of metformin. In addition, these data identify induction of hepatic GCN5 as a potential therapeutic strategy for treatment of T2DM.


Diabetes, Obesity and Metabolism | 2011

Metformin opposes impaired AMPK and SIRT1 function and deleterious changes in core clock protein expression in white adipose tissue of genetically-obese db/db mice.

Paul Caton; Julius Kieswich; Muhammad M. Yaqoob; Mark J. Holness; Mary C. Sugden

Aim: AMPK activates SIRT1 in liver and skeletal muscle. Impaired circadian function is associated with development of obesity. SIRT1 regulates circadian function and is suppressed in white adipose tissue (WAT) of obese patients. We examined the potential role of AMPK and SIRT1 in regulation of circadian components in WAT of obese db/db mice and in mice fed a high‐fat diet (HFD), and investigated whether metformin‐mediated activation of AMPK opposed any deleterious changes in the WAT clock mechanism.


Molecular and Cellular Biology | 2004

Nuclear Speckle-Associated Protein Pnn/DRS Binds to the Transcriptional Corepressor CtBP and Relieves CtBP- Mediated Repression of the E-Cadherin Gene

Roman Alpatov; Gustavo C. Munguba; Paul Caton; Jeong-Hoon Joo; Yang Shi; Me Hunt; Stephen P. Sugrue

ABSTRACT Previously, we have shown that pinin/DRS (Pnn), a 140-kDa nuclear and cell adhesion-related phosphoprotein, is involved in the regulation of cell adhesion and modulation of the activity of multiple tumor suppressor genes. In the nucleus Pnn is concentrated in the “nuclear speckles,” zones of accumulation of transcriptional and mRNA splicing factors, where Pnn is involved in mRNA processing. Alternatively, other roles of Pnn in gene regulation have not yet been established. By utilizing in vitro pull-down assays, in vivo interaction studies, and immunofluorescence in combination with overexpression and RNA interference experiments, we present evidence that Pnn interacts with the known transcriptional corepressor CtBP1. As a consequence of this interaction Pnn was capable of relieving the CtBP1-mediated repression of E-cadherin promoter activity. Our results suggest that the interaction of Pnn with the corepressor CtBP1 may modulate repression of transcription by CtBP1. This interaction may reflect the existence of coupling factors involved in CtBP-mediated transcriptional regulation and mRNA processing events.


Diabetologia | 2013

Sirtuin 3 regulates mouse pancreatic beta cell function and is suppressed in pancreatic islets isolated from human type 2 diabetic patients

Paul Caton; Sarah J. Richardson; Julius Kieswich; Marco Bugliani; Michelle L. Holland; Piero Marchetti; Noel G. Morgan; Muhammad M. Yaqoob; Mark J. Holness; Mary C. Sugden

Aims/hypothesisSirtuin (SIRT)3 is a mitochondrial protein deacetylase that regulates reactive oxygen species (ROS) production and exerts anti-inflammatory effects. As chronic inflammation and mitochondrial dysfunction are key factors mediating pancreatic beta cell impairment in type 2 diabetes, we investigated the role of SIRT3 in the maintenance of beta cell function and mass in type 2 diabetes.MethodsWe analysed changes in SIRT3 expression in experimental models of type 2 diabetes and in human islets isolated from type 2 diabetic patients. We also determined the effects of SIRT3 knockdown on beta cell function and mass in INS1 cells.ResultsSIRT3 expression was markedly decreased in islets isolated from type 2 diabetes patients, as well as in mouse islets or INS1 cells incubated with IL1β and TNFα. SIRT3 knockdown in INS1 cells resulted in lowered insulin secretion, increased beta cell apoptosis and reduced expression of key beta cell genes. SIRT3 knockdown also blocked the protective effects of nicotinamide mononucleotide on pro-inflammatory cytokines in beta cells. The deleterious effects of SIRT3 knockdown were mediated by increased levels of cellular ROS and IL1β.Conclusions/interpretationDecreased beta cell SIRT3 levels could be a key step in the onset of beta cell dysfunction, occurring via abnormal elevation of ROS levels and amplification of beta cell IL1β synthesis. Strategies to increase the activity or levels of SIRT3 could generate attractive therapies for type 2 diabetes.


Journal of Agricultural and Food Chemistry | 2010

Regulation of Vascular Endothelial Function by Procyanidin-Rich Foods and Beverages†

Paul Caton; Mark R. Pothecary; Delphine M. Lees; Noorafza Q. Khan; Elizabeth G. Wood; Toshihiko Shoji; Tomomasa Kanda; Gurvinder Rull; Roger Corder

Flavonoid-rich diets are associated with a lower mortality from cardiovascular disease. This has been linked to improvements in endothelial function. However, the specific flavonoids, or biologically active metabolites, conferring these beneficial effects have yet to be fully defined. In this experimental study of the effect of flavonoids on endothelial function cultured endothelial cells have been used as a bioassay with endothelin-1 (ET-1) synthesis being measured an index of the response. Evaluation of the relative effects of extracts of cranberry juice compared to apple, cocoa, red wine, and green tea showed inhibition of ET-1 synthesis was dependent primarily on their oligomeric procyanidin content. Procyanidin-rich extracts of cranberry juice triggered morphological changes in endothelial cells with reorganization of the actin cytoskeleton and increased immunostaining for phosphotyrosine residues. These actions were independent of antioxidant activity. Comparison of the effects of apple procyanidin monomers through heptamer showed a clear structure-activity relationship. Although monomer, dimer, and trimer had little effect on ET-1 synthesis, procyanidin tetramer, pentamer, hexamer, and heptamer produced concentration-dependent decreases with IC(50) values of 5.4, 1.6, 0.9, and 0.7 microM, respectively. Levels of ET-1 mRNA showed a similar pattern of decreases, which were inversely correlated with increased expression of Kruppel-like factor 2 (KLF2), a key endothelial transcription factor with a broad range of antiatherosclerotic actions including suppression of ET-1 synthesis. Future investigations of procyanidin-rich products should assess the role KLF2 induction plays in the beneficial vascular effects of high flavonoid consumption.


Journal of Endocrinology | 2011

Fructose induces gluconeogenesis and lipogenesis through a SIRT1-dependent mechanism

Paul Caton; Nanda Nayuni; Noorafza Q. Khan; Elizabeth G. Wood; Roger Corder

Consumption of a fructose-rich diet leads to insulin resistance and dyslipidemia in part due to elevated gluconeogenesis and lipogenesis. SIRT1, an NAD(+)-dependent protein deacetylase, can induce gluconeogenesis and lipogenesis. The aim of this study was to determine whether fructose increased hepatic SIRT1, leading to induction of gluconeogenesis and lipogenesis. Rat hepatocytes were incubated with fructose (1-5 mM). SIRT1 protein, SIRT1 activity, and NAD(+)/NADH ratio were measured. The effects of SIRT1 inhibitors (EX-527 and nicotinamide) and activators (SIRT1 activator 3 and SRT1720) and the mitochondrial complex I inhibitor rotenone were examined on fructose-induced increases in gluconeogenesis and lipogenesis. Fructose increased SIRT1 protein, SIRT1 activity, and NAD(+)/NADH ratio. Fructose also induced gluconeogenesis, with increases in peroxisome proliferator-activated receptor coactivator 1-alpha (PGC1α) and phosphoenolpyruvate carboxykinase (PEPCK; gene code Pck1) gene expression, PEPCK activity, and hepatocyte glucose production. In addition, levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase (Hmgcr) and acetyl-coA carboxylase (Acc) mRNA, and intracellular cholesterol were increased. Increases in gluconeogenesis, Hmgcr, Acc, and cholesterol were abolished by SIRT1 inhibitors and rotenone, while SIRT1 activators increased gluconeogenesis, Hmgcr, Acc, Pgc1β, and sterol regulatory element-binding protein 1c (Srebp1c) gene expression. In conclusion, fructose induces gluconeogenesis and lipogenesis through a SIRT1-dependent mechanism, suggesting that induction of hepatic SIRT1 could play a pivotal role in the metabolic changes observed in humans and animals consuming a fructose-rich diet. These results highlight the need for a greater understanding of the role of SIRT1 in metabolic regulation and indicate the potential for adverse effects of SIRT1 activators if used therapeutically.


Nutrition | 2010

Acute and long-term nutrient-led modifications of gene expression: Potential role of SIRT1 as a central co-ordinator of short and longer-term programming of tissue function

Mark J. Holness; Paul Caton; Mary C. Sugden

Environmental factors can influence the acute and longer-term risks of developing diseases, including type 2 diabetes mellitus and cardiovascular disease; however, the underlying mechanism remains elusive. Increasing evidence suggests that these effects can be achieved by modification of metabolic gene expression. These include acute changes in histone methylation, acetylation, phosphorylation, and ubiquitination and longer-term DNA silencing elicited by DNA methylation. Thus, an increased risk of disease may reflect acute or chronic stable modification of genes that regulate nutrient handling, leading to altered nutrient utilization (increased lipid oxidation at the expense of glucose utilization) and/or changes in the balance between nutrient storage and energy production, thereby favoring the development of obesity. The review addresses the hypothesis that early-life epigenetic programming of gene expression could be mirrored by changes in acute function of nuclear receptors, in particular the peroxisome proliferator-activated receptors, achieved by enzymes that are more conventionally involved in regulating DNA methylation and post-transcriptional modification of histones. Emphasis is placed on the potential importance of the protein deacetylase sirtuin-1 as a central co-ordinator.


Biochemical Journal | 2011

PPARα-LXR as a novel metabolostatic signalling axis in skeletal muscle that acts to optimize substrate selection in response to nutrient status

Paul Caton; Mark J. Holness; David Bishop-Bailey; Mary C. Sugden

LXR (liver X receptor) and PPARα (peroxisome-proliferator-activated receptor α) are nuclear receptors that control the expression of genes involved in glucose and lipid homoeostasis. Using wild-type and PPARα-null mice fed on an LXR-agonist-supplemented diet, the present study analysed the impact of pharmacological LXR activation on the expression of metabolically important genes in skeletal muscle, testing the hypothesis that LXR activation can modulate PPAR action in skeletal muscle in a manner dependent on nutritional status. In the fed state, LXR activation promoted a gene profile favouring lipid storage and glucose oxidation, increasing SCD1 (stearoyl-CoA desaturase 1) expression and down-regulating PGC-1α (PPARγ co-activator-1α) and PDK4 (pyruvate dehydrogenase kinase 4) expression. PPARα deficiency enhanced LXR stimulation of SCD1 expression, and facilitated elevated SREBP-1 (sterol-regulatory-element-binding protein-1) expression. However, LXR-mediated down-regulation of PGC-1α and PDK4 was opposed and reversed by PPARα deficiency. During fasting, prior LXR activation augmented PPARα signalling to heighten FA (fatty acid) oxidation and decrease glucose oxidation by augmenting fasting-induced up-regulation of PGC-1α and PDK4 expression, effects opposed by PPARα deficiency. Starvation-induced down-regulation of SCD1 expression was opposed by antecedent LXR activation in wild-type mice, an effect enhanced further by PPARα deficiency, which may elicit increased channelling of FA into triacylglycerol to limit lipotoxicity. Our results also identified potential regulatory links between the protein deacetylases SIRT1 (sirtuin 1) and SIRT3 and PDK4 expression in muscle from fasted mice, with a requirement for PPARα. In summary, we therefore propose that a LXR-PPARα signalling axis acts as a metabolostatic regulatory mechanism to optimize substrate selection and disposition in skeletal muscle according to metabolic requirement.


Science | 2016

Early-life nutrition modulates the epigenetic state of specific rDNA genetic variants in mice

Michelle L. Holland; Robert Lowe; Paul Caton; Carolina Gemma; Guillermo Carbajosa; Amy F. Danson; Asha A. M. Carpenter; Elena Loche; Susan E. Ozanne; Vardhman K. Rakyan

Moms diet affects growth Nutrition is important during development. This appears to be true even in utero, with potential long-lasting effects on adult phenotype and disease. Epigenetic factors are prime suspects in identifying the corresponding molecular mechanism because they can be maintained throughout cell division. Holland et al. show in mice that in utero nutritional deficits influence offspring growth through epigenetic states at multicopy ribosomal DNA elements. This effect is influenced by the genetic variation that naturally exists within the ribosomal RNA. Science, this issue p. 495 Maternal environment induces epigenetic dynamics at ribosomal DNA in the mouse. A suboptimal early-life environment, due to poor nutrition or stress during pregnancy, can influence lifelong phenotypes in the progeny. Epigenetic factors are thought to be key mediators of these effects. We show that protein restriction in mice from conception until weaning induces a linear correlation between growth restriction and DNA methylation at ribosomal DNA (rDNA). This epigenetic response remains into adulthood and is restricted to rDNA copies associated with a specific genetic variant within the promoter. Related effects are also found in models of maternal high-fat or obesogenic diets. Our work identifies environmentally induced epigenetic dynamics that are dependent on underlying genetic variation and establishes rDNA as a genomic target of nutritional insults.

Collaboration


Dive into the Paul Caton's collaboration.

Top Co-Authors

Avatar

Julius Kieswich

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Mary C. Sugden

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Mark J. Holness

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Muhammad M. Yaqoob

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Roger Corder

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Nanda Nayuni

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katie Hanna

Nottingham Trent University

View shared research outputs
Top Co-Authors

Avatar

Noorafza Q. Khan

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge