Network


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

Hotspot


Dive into the research topics where Timothy J. Pullen is active.

Publication


Featured researches published by Timothy J. Pullen.


Molecular and Cellular Biology | 2011

miR-29a and miR-29b Contribute to Pancreatic β-Cell-Specific Silencing of Monocarboxylate Transporter 1 (Mct1)

Timothy J. Pullen; Gabriela da Silva Xavier; Gavin Kelsey; Guy A. Rutter

ABSTRACT In pancreatic β cells, elevated glucose concentrations stimulate mitochondrial oxidative metabolism to raise intracellular ATP/ADP levels, prompting insulin secretion. Unusually low levels of expression of genes encoding the plasma membrane monocarboxylate transporter, MCT1 (SLC16A1), as well as lactate dehydrogenase A (LDHA) ensure that glucose-derived pyruvate is efficiently metabolized by mitochondria, while exogenous lactate or pyruvate is unable to stimulate metabolism and hence insulin secretion inappropriately. We show here that whereas DNA methylation at the Mct1 promoter is unlikely to be involved in cell-type-specific transcriptional repression, three microRNAs (miRNAs), miR-29a, miR-29b, and miR-124, selectively target both human and mouse MCT1 3′ untranslated regions. Mutation of the cognate miR-29 or miR-124 binding sites abolishes the effects of the corresponding miRNAs, demonstrating a direct action of these miRNAs on the MCT1 message. However, despite reports of its expression in the mouse β-cell line MIN6, miR-124 was not detectably expressed in mature mouse islets. In contrast, the three isoforms of miR-29 are highly expressed and enriched in mouse islets. We show that inhibition of miR-29a in primary mouse islets increases Mct1 mRNA levels, demonstrating that miR-29 isoforms contribute to the β-cell-specific silencing of the MCT1 transporter and may thus affect insulin release.


PLOS ONE | 2012

The mitochondrial Ca2+ uniporter MCU is essential for glucose-induced ATP increases in pancreatic β-cells.

Andrei I. Tarasov; Francesca Semplici; Magalie A. Ravier; Elisa A. Bellomo; Timothy J. Pullen; Patrick Gilon; Israel Sekler; Rosario Rizzuto; Guy A. Rutter

Glucose induces insulin release from pancreatic β-cells by stimulating ATP synthesis, membrane depolarisation and Ca2+ influx. As well as activating ATP-consuming processes, cytosolic Ca2+ increases may also potentiate mitochondrial ATP synthesis. Until recently, the ability to study the role of mitochondrial Ca2+ transport in glucose-stimulated insulin secretion has been hindered by the absence of suitable approaches either to suppress Ca2+ uptake into these organelles, or to examine the impact on β-cell excitability. Here, we have combined patch-clamp electrophysiology with simultaneous real-time imaging of compartmentalised changes in Ca2+ and ATP/ADP ratio in single primary mouse β-cells, using recombinant targeted (Pericam or Perceval, respectively) as well as entrapped intracellular (Fura-Red), probes. Through shRNA-mediated silencing we show that the recently-identified mitochondrial Ca2+ uniporter, MCU, is required for depolarisation-induced mitochondrial Ca2+ increases, and for a sustained increase in cytosolic ATP/ADP ratio. By contrast, silencing of the mitochondrial Na+-Ca2+ exchanger NCLX affected the kinetics of glucose-induced changes in, but not steady state values of, cytosolic ATP/ADP. Exposure to gluco-lipotoxic conditions delayed both mitochondrial Ca2+ uptake and cytosolic ATP/ADP ratio increases without affecting the expression of either gene. Mitochondrial Ca2+ accumulation, mediated by MCU and modulated by NCLX, is thus required for normal glucose sensing by pancreatic β-cells, and becomes defective in conditions mimicking the diabetic milieu.


Islets | 2010

Identification of genes selectively disallowed in the pancreatic islet

Timothy J. Pullen; Arshad M. Khan; Geraint Barton; Sarah Butcher; Gao Sun; Guy A. Rutter

We have previously identified two genes, encoding lactate dehydrogenase (Ldha) and the monocarboxylate carrier, MCT1 (Slc16a1) whose expression is remarkably low in pancreatic β-cells and islets. We sought here to determine whether these may be part of a larger family of genes selectively repressed (“disallowed”) in the pancreatic islet. Using new and publicly available microarray data, we undertook a bioinformatic analysis of gene expression in islets and a range of other murine tissues. We compared data sets from three sources of mouse pancreatic islets with a total of 30 datasets from nine tissues, to identify genes with at least five-fold down-regulation in islets. 39 genes were revealed as being specifically repressed in islets. These included Ldha and Slc16a1 as expected but also genes involved in several other metabolic pathways which could affect glucose stimulated insulin secretion. Of these, adenylate kinase 3 (AK3) is a mitochondrial enzyme which acts on GTP, and ornithine aminotransferase (OAT) lies on the pathway converting glutamate to ornithine. The removal of an enzyme which could dissipate mitochondrial GTP levels in beta cells provides support for the theory that mitochondrial GTP may be an important for regulating insulin secretion, whilst blocking an alternative metabolic fate for glutamate is consistent with a signalling role for glutamate. The identification of these genes should inform efforts to generate fully functional β-cells from stem cell sources, and may provide new targets in type 2 diabetes.


Diabetes | 2010

Carbohydrate-Responsive Element-Binding Protein (ChREBP) Is a Negative Regulator of ARNT/HIF-1β Gene Expression in Pancreatic Islet β-Cells

Nafeesa A. Noordeen; Tarnjit K. Khera; Gao Sun; E. Rebecca Longbottom; Timothy J. Pullen; Gabriela da Silva Xavier; Guy A. Rutter; Isabelle Leclerc

OBJECTIVE Carbohydrate-responsive element-binding protein (ChREBP) is a transcription factor that has been shown to regulate carbohydrate metabolism in the liver and pancreatic β-cells in response to elevated glucose concentrations. Because few genes have been identified so far as bona fide ChREBP-target genes, we have performed a genome-wide analysis of the ChREBP transcriptome in pancreatic β-cells. RESEARCH DESIGN AND METHODS Chromatin immunoprecipitation and high-density oligonucleotide tiling arrays (ChIP-chip; Agilent Technologies) using MIN6 pancreatic β-cell extracts were performed together with transcriptional and other analysis using standard techniques. RESULTS One of the genes identified by ChIP-chip and linked to glucose sensing and insulin secretion was aryl hydrocarbon receptor nuclear translocator (ARNT)/hypoxia-inducible factor-1β (HIF-1β), a transcription factor implicated in altered gene expression and pancreatic-islet dysfunction in type 2 diabetes. We first confirmed that elevated glucose concentrations decreased ARNT/HIF-1β levels in INS-1 (832/13) cells and primary mouse islets. Demonstrating a role for ChREBP in ARNT gene regulation, ChREBP silencing increased ARNT mRNA levels in INS-1 (832/13) cells, and ChREBP overexpression decreased ARNT mRNA in INS-1 (832/13) cells and primary mouse islets. We demonstrated that ChREBP and Max-like protein X (MLX) bind on the ARNT/HIF-1β promoter on the proximal region that also confers the negative glucose responsiveness. CONCLUSIONS These results demonstrate that ChREBP acts as a novel repressor of the ARNT/HIF-1β gene and might contribute to β-cell dysfunction induced by glucotoxicity.


Diabetes | 2014

ADCY5 Couples Glucose to Insulin Secretion in Human Islets

David J. Hodson; Ryan K. Mitchell; Lorella Marselli; Timothy J. Pullen; Silvia Gimeno Brias; Francesca Semplici; Katy L. Everett; Dermot M. F. Cooper; Marco Bugliani; Piero Marchetti; Vanessa Lavallard; Domenico Bosco; Lorenzo Piemonti; Paul Johnson; Stephen J. Hughes; Daliang Li; Wen Hong Li; A. M. James Shapiro; Guy A. Rutter

Single nucleotide polymorphisms (SNPs) within the ADCY5 gene, encoding adenylate cyclase 5, are associated with elevated fasting glucose and increased type 2 diabetes (T2D) risk. Despite this, the mechanisms underlying the effects of these polymorphic variants at the level of pancreatic β-cells remain unclear. Here, we show firstly that ADCY5 mRNA expression in islets is lowered by the possession of risk alleles at rs11708067. Next, we demonstrate that ADCY5 is indispensable for coupling glucose, but not GLP-1, to insulin secretion in human islets. Assessed by in situ imaging of recombinant probes, ADCY5 silencing impaired glucose-induced cAMP increases and blocked glucose metabolism toward ATP at concentrations of the sugar >8 mmol/L. However, calcium transient generation and functional connectivity between individual human β-cells were sharply inhibited at all glucose concentrations tested, implying additional, metabolism-independent roles for ADCY5. In contrast, calcium rises were unaffected in ADCY5-depleted islets exposed to GLP-1. Alterations in β-cell ADCY5 expression and impaired glucose signaling thus provide a likely route through which ADCY5 gene polymorphisms influence fasting glucose levels and T2D risk, while exerting more minor effects on incretin action.


Diabetes, Obesity and Metabolism | 2013

When less is more: the forbidden fruits of gene repression in the adult β‐cell

Timothy J. Pullen; Guy A. Rutter

Outside of the biological arena the term ‘repression’ often has a negative connotation. However, in the pancreatic β‐cell a small group of genes, which are abundantly expressed in most if not all other mammalian tissues, are highly selectively repressed, with likely functional consequences. The two ‘founder’ members of this group, lactate dehydrogenase A (Ldha) and monocarboxylate transporter‐1 (MCT‐1/Slc16a1), are inactivated by multiple mechanisms including histone modifications and microRNA‐mediated silencing. Their inactivation ensures that pyruvate and lactate, derived from muscle during exercise, do not stimulate insulin release inappropriately. Correspondingly, activating mutations in the MCT‐1 promoter underlie ‘exercise‐induced hyperinsulinism’ (EIHI) in man, a condition mimicked by forced over‐expression of MCT‐1 in the β‐cell in mice. Furthermore, LDHA expression in the β‐cell is upregulated in both human type 2 diabetes and in rodent models of the disease. Recent work by us and by others has identified a further ∼60 genes which are selectively inactivated in the β‐cell, a list which we refine here up to seven by detailed comparison of the two studies. These genes include key regulators of cell proliferation and stimulus‐secretion coupling. The present, and our earlier results, thus highlight the probable importance of shutting down a subset of ‘disallowed’ genes for the differentiated function of β‐cells, and implicate previously unsuspected signalling pathways in the control of β‐cell expansion and insulin secretion. Targeting of deregulated ‘disallowed’ genes in these cells may thus, in the future, provide new therapeutic avenues for type 2 diabetes.


Diabetes | 2012

Overexpression of Monocarboxylate Transporter-1 (Slc16a1) in Mouse Pancreatic β-Cells Leads to Relative Hyperinsulinism During Exercise

Timothy J. Pullen; Lykke Sylow; Gao Sun; Andrew P. Halestrap; Erik A. Richter; Guy A. Rutter

Exercise-induced hyperinsulinism (EIHI) is an autosomal dominant disorder characterized by inappropriate insulin secretion in response to vigorous physical exercise or pyruvate injection. Activating mutations in the monocarboxylate transporter-1 (MCT1, SLC16A1) promoter have been linked to EIHI. Expression of this pyruvate transporter is specifically repressed (disallowed) in pancreatic β-cells, despite nearly universal expression across other tissues. It has been impossible to determine, however, whether EIHI mutations cause MCT1 expression in patient β-cells. The hypothesis that MCT1 expression in β-cells is sufficient to cause EIHI by allowing entry of pyruvate and triggering insulin secretion thus remains unproven. Therefore, we generated a transgenic mouse capable of doxycycline-induced, β-cell–specific overexpression of MCT1 to test this model directly. MCT1 expression caused isolated islets to secrete insulin in response to pyruvate, without affecting glucose-stimulated insulin secretion. In vivo, transgene induction lowered fasting blood glucose, mimicking EIHI. Pyruvate challenge stimulated increased plasma insulin and smaller excursions in blood glucose in transgenic mice. Finally, in response to exercise, transgene induction prevented the normal inhibition of insulin secretion. Forced overexpression of MCT1 in β-cells thus replicates the key features of EIHI and highlights the importance of this transporter’s absence from these cells for the normal control of insulin secretion.


The FASEB Journal | 2014

LKB1 and AMPK differentially regulate pancreatic β-cell identity

Marina Kone; Timothy J. Pullen; Gao Sun; Mark Ibberson; Aida Martinez-Sanchez; Sophie Sayers; Marie-Sophie Nguyen-Tu; Chase Kantor; Avital Swisa; Yuval Dor; Tracy Gorman; Jorge Ferrer; Bernard Thorens; Frank Reimann; Fiona M. Gribble; James McGinty; Lingling Chen; Paul M. W. French; Fabian Birzele; Tobias Hildebrandt; Ingo Uphues; Guy A. Rutter

Fully differentiated pancreatic β cells are essential for normal glucose homeostasis in mammals. Dedifferentiation of these cells has been suggested to occur in type 2 diabetes, impairing insulin production. Since chronic fuel excess (“glucotoxicity”) is implicated in this process, we sought here to identify the potential roles in β‐cell identity of the tumor suppressor liver kinase B1 (LKB1/STK11) and the downstream fuel‐sensitive kinase, AMP‐activated protein kinase (AMPK). Highly β‐cell‐restricted deletion of each kinase in mice, using an Ins1‐controlled Cre, was therefore followed by physiological, morphometric, and massive parallel sequencing analysis. Loss of LKB1 strikingly (2.0‐12‐fold, E<0.01) increased the expression of subsets of hepatic (Alb, Iyd, Elovl2) and neuronal (Nptx2, Dlgap2, Cartpt, Pdyn) genes, enhancing glutamate signaling. These changes were partially recapitulated by the loss of AMPK, which also up‐regulated β‐cell “disallowed” genes (Slc16a1, Ldha, Mgst1, Pdgfra) 1.8‐ to 3.4‐fold (E<0.01). Correspondingly, targeted promoters were enriched for neuronal (Zfp206; P= 1.3×10‐33) and hypoxia‐regulated (HIF1; P= 2.5×10‐16) transcription factors. In summary, LKB1 and AMPK, through only partly overlapping mechanisms, maintain β‐cell identity by suppressing alternate pathways leading to neuronal, hepatic, and other characteristics. Selective targeting of these enzymes may provide a new approach to maintaining β‐cell function in some forms of diabetes.—Kone, M., Pullen, T. J., Sun, G., Ibberson, M., Martinez‐Sanchez, A., Sayers, S., Nguyen‐Tu, M.‐S., Kantor, C., Swisa, A., Dor, Y., Gorman, T., Ferrer, J., Thorens, B., Reimann, F., Gribble, F., McGinty, J. A., Chen, L., French, P. M., Birzele, F., Hildebrandt, T., Uphues, I., Rutter, G. A., LKB1 and AMPK differentially regulate pancreatic β‐cell identity. FASEB J. 28, 4972–4985 (2014). www.fasebj.org


Cell Metabolism | 2016

Chronic Activation of γ2 AMPK Induces Obesity and Reduces β Cell Function

Arash Yavari; Claire J. Stocker; Sahar Ghaffari; Edward T. Wargent; Violetta Steeples; Gabor Czibik; Katalin Pinter; Mohamed Bellahcene; Angela Woods; Pablo Blanco Martinez de Morentin; Celine Cansell; Brian Yee Hong Lam; André Chuster; Kasparas Petkevicius; Marie-Sophie Nguyen-Tu; Aida Martinez-Sanchez; Timothy J. Pullen; Peter L. Oliver; A Stockenhuber; Chinh Nguyen; Merzaka Lazdam; Jacqueline F. O’Dowd; Parvathy E. Harikumar; Mónika Tóth; Craig Beall; Theodosios Kyriakou; Julia Parnis; Dhruv Sarma; George Katritsis; Diana D.J. Wortmann

Summary Despite significant advances in our understanding of the biology determining systemic energy homeostasis, the treatment of obesity remains a medical challenge. Activation of AMP-activated protein kinase (AMPK) has been proposed as an attractive strategy for the treatment of obesity and its complications. AMPK is a conserved, ubiquitously expressed, heterotrimeric serine/threonine kinase whose short-term activation has multiple beneficial metabolic effects. Whether these translate into long-term benefits for obesity and its complications is unknown. Here, we observe that mice with chronic AMPK activation, resulting from mutation of the AMPK γ2 subunit, exhibit ghrelin signaling-dependent hyperphagia, obesity, and impaired pancreatic islet insulin secretion. Humans bearing the homologous mutation manifest a congruent phenotype. Our studies highlight that long-term AMPK activation throughout all tissues can have adverse metabolic consequences, with implications for pharmacological strategies seeking to chronically activate AMPK systemically to treat metabolic disease.


Journal of Biological Chemistry | 2011

Nucleo-cytosolic shuttling of FoxO1 directly regulates mouse Ins2 but not Ins1 gene expression in pancreatic beta cells (MIN6).

Gargi Meur; Qingwen Qian; Gabriela da Silva Xavier; Timothy J. Pullen; Takashi Tsuboi; Caroline McKinnon; Lm Fletcher; Jeremy M. Tavaré; Stephen H. Hughes; Paul Johnson; Guy A. Rutter

The Forkhead box transcription factor FoxO1 regulates metabolic gene expression in mammals. FoxO1 activity is tightly controlled by phosphatidylinositol 3-kinase (PI3K) signaling, resulting in its phosphorylation and nuclear exclusion. We sought here to determine the mechanisms involved in glucose and insulin-stimulated nuclear shuttling of FoxO1 in pancreatic β cells and its consequences for preproinsulin (Ins1, Ins2) gene expression. Nuclear-localized endogenous FoxO1 translocated to the cytosol in response to elevated glucose (3 versus 16.7 mm) in human islet β cells. Real-time confocal imaging of nucleo-cytosolic shuttling of a FoxO1-EGFP chimera in primary mouse and clonal MIN6 β cells revealed a time-dependent glucose-responsive nuclear export, also mimicked by exogenous insulin, and blocked by suppressing insulin secretion. Constitutively active PI3K or protein kinase B/Akt exerted similar effects, while inhibitors of PI3K, but not of glycogen synthase kinase-3 or p70 S6 kinase, blocked nuclear export. FoxO1 overexpression reversed the activation by glucose of pancreatic duodenum homeobox-1 (Pdx1) transcription. Silencing of FoxO1 significantly elevated the expression of mouse Ins2, but not Ins1, mRNA at 3 mm glucose. Putative FoxO1 binding sites were identified in the distal promoter of rodent Ins2 genes and direct binding of FoxO1 to the Ins2 promoter was demonstrated by chromatin immunoprecipitation. A 915-bp glucose-responsive Ins2 promoter was inhibited by constitutively active FoxO1, an effect unaltered by simultaneous overexpression of PDX1. We conclude that nuclear import of FoxO1 contributes to the suppression of Pdx1 and Ins2 gene expression at low glucose, the latter via a previously unsuspected and direct physical interaction with the Ins2 promoter.

Collaboration


Dive into the Timothy J. Pullen's collaboration.

Top Co-Authors

Avatar

Guy A. Rutter

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Gao Sun

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabelle Leclerc

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge