Network


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

Hotspot


Dive into the research topics where Accalia Fu is active.

Publication


Featured researches published by Accalia Fu.


Cell Metabolism | 2009

Loss of Lkb1 in Adult β Cells Increases β Cell Mass and Enhances Glucose Tolerance in Mice

Accalia Fu; Andy Cheuk-Him Ng; Chantal Depatie; Nadeeja Wijesekara; Ying He; Gen-Sheng Wang; Nabeel Bardeesy; Fraser W. Scott; Rhian M. Touyz; Michael B. Wheeler; Robert A. Screaton

The Lkb1 tumor suppressor exerts its biological effects through phosphorylation and consequent activation of the AMP kinase (AMPK) family. Extensive genetic and biochemical evidence supports a role for Lkb1 in cell cycle arrest, establishment of cell polarity, and cellular energy metabolism. However, the role of Lkb1 and the AMPK family in beta cell function in vivo has not been established. We generated conditional knockout mice with a deletion of the Lkb1 gene in the beta cell compartment of pancreatic islets; these mice display improved glucose tolerance and protection against diet-induced hyperglycemia. Lkb1(-/-) beta cells are hypertrophic because of elevated mTOR activity; they also proliferate more and secrete more insulin in response to glucose. These data indicate that inhibiting Lkb1 activity in beta cells may facilitate beta cell expansion and glucose tolerance in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Glucose controls CREB activity in islet cells via regulated phosphorylation of TORC2

Deidre Jansson; Andy Cheuk-Him Ng; Accalia Fu; Chantal Depatie; Mufida Al Azzabi; Robert A. Screaton

CREB is a cAMP- and calcium-responsive transcriptional activator that is required for islet beta cell proliferation and survival. Glucose and incretin hormones elicit beta cell insulin secretion and promote synergistic CREB activity by inducing the nuclear relocalization of TORC2 (also known as Crtc2), a coactivator for CREB. In islet cells under basal conditions when CREB activity is low, TORC2 is phosphorylated and sequestered in the cytoplasm by 14-3-3 proteins. In response to feeding stimuli, TORC2 is dephosphorylated, enters the nucleus, and binds to CREB located at target gene promoters. The dephosphorylation of TORC2 at Ser-171 in response to cAMP is insufficient to account for the dynamics of TORC2 localization and CREB activity in islet cells. Here, we identify Ser-275 of TORC2 as a 14-3-3 binding site that is phosphorylated under low glucose conditions and which becomes dephosphorylated by calcineurin in response to glucose influx. Dephosphorylation of Ser-275 is essential for both glucose and cAMP-mediated activation of CREB in beta cells and islets. Using a cell-based screen of 180 human protein kinases, we identified MARK2, a member of the AMPK family of Ser/Thr kinases, as a Ser-275 kinase that blocks TORC2:CREB activity. Taken together, these data provide the mechanistic underpinning for how cAMP and glucose cooperatively promote a transcriptional program critical for islet cell survival, and identifies MARK2 as a potential target for diabetes treatment.


Molecular and Cellular Endocrinology | 2013

Role of AMPK in pancreatic beta cell function

Accalia Fu; Chandra E. Eberhard; Robert A. Screaton

Pharmacological activation of AMP activated kinase (AMPK) by metformin has proven to be a beneficial therapeutic approach for the treatment of type II diabetes. Despite improved glucose regulation achieved by administration of small molecule activators of AMPK, the potential negative impact of enhanced AMPK activity on insulin secretion by the pancreatic beta cell is an important consideration. In this review, we discuss our current understanding of the role of AMPK in central functions of the pancreatic beta cell, including glucose-stimulated insulin secretion (GSIS), proliferation, and survival. In addition we discuss the controversy surrounding the role of AMPK in insulin secretion, underscoring the merits and caveats of methods used to date.


Journal of Biological Chemistry | 2012

Glutathionylation state of uncoupling protein-2 and the control of glucose-stimulated insulin secretion.

Ryan J. Mailloux; Accalia Fu; Christine A. Robson-Doucette; Emma M. Allister; Michael B. Wheeler; Robert A. Screaton; Mary-Ellen Harper

Background: Proton leak through UCP2 is modulated by glutathionylation, and UCP2 modulates GSIS. Results: Glutathionylation of UCP2 amplifies GSIS from β cells. Matrix ROS activates UCP2-desensitizing GSIS. Conclusion: Reversible glutathionylation of UCP2 aids in regulating GSIS. Significance: Findings enhance our understanding of the role of redox circuits in the modulation of GSIS. The role of reactive oxygen species (ROS) in glucose-stimulated insulin release remains controversial because ROS have been shown to both amplify and impede insulin release. In regard to preventing insulin release, ROS activates uncoupling protein-2 (UCP2), a mitochondrial inner membrane protein that negatively regulates glucose-stimulated insulin secretion (GSIS) by uncoupling oxidative phosphorylation. With our recent discovery that the UCP2-mediated proton leak is modulated by reversible glutathionylation, a process responsive to small changes in ROS levels, we resolved to determine whether glutathionylation is required for UCP2 regulation of GSIS. Using Min6 cells and pancreatic islets, we demonstrate that induction of glutathionylation not only deactivates UCP2-mediated proton leak but also enhances GSIS. Conversely, an increase in mitochondrial matrix ROS was found to deglutathionylate and activate UCP2 leak and impede GSIS. Glucose metabolism also decreased the total amount of cellular glutathionylated proteins and increased the cellular glutathione redox ratio (GSH/GSSG). Intriguingly, the provision of extracellular ROS (H2O2, 10 μm) amplified GSIS and also activated UCP2. Collectively, our findings indicate that the glutathionylation status of UCP2 contributes to the regulation of GSIS, and different cellular sites and inducers of ROS can have opposing effects on GSIS, perhaps explaining some of the controversy surrounding the role of ROS in GSIS.


Endocrinology | 2013

CRTC2 Is Required for β-Cell Function and Proliferation

Chandra E. Eberhard; Accalia Fu; Courtney Reeks; Robert A. Screaton

Previous work in insulinoma cell lines has established that calcineurin plays a critical role in the activation of cAMP-responsive element binding protein (Creb), a key transcription factor required for β-cell function and survival, by dephosphorylating the Creb coactivator Creb-regulated transcription coactivator (Crtc)2 at 2 regulatory sites, Ser171 and Ser275. Here, we report that Crtc2 is essential both for glucose-stimulated insulin secretion and cell survival in the β-cell. Endogenous Crtc2 activation is achieved via increasing glucose levels to the physiological feeding range, indicating that Crtc2 is a sensor that couples ambient glucose concentrations to Creb activity in the β-cell. Immunosuppressant drugs such as cyclosporin A and tacrolimus that target the protein phosphatase calcineurin are commonly administered after organ transplantation. Chronic use is associated with reduced insulin secretion and new onset diabetes, suggestive of pancreatic β-cell dysfunction. Importantly, we show that overexpression of a Crtc2 mutant rendered constitutively active by introduction of nonphosphorylatable alanine residues at Ser171 and Ser275 permits Creb target gene activation under conditions when calcineurin is inhibited. Taken together, these data suggest that promoting Crtc2-Creb activity is required for β-cell function and proliferation and promoting this pathway could ameliorate symptoms of new onset diabetes after transplantation.


Cell Cycle | 2008

Using kinomics to delineate signaling pathways: Control of crtc2/torc2 by the ampk family

Accalia Fu; Robert A. Screaton

The classical role of AMP-activated protein kinase (AMPK) as an energy status sensor is expanding to include other members of the AMPK family. Recent genetic and cell biological evidence points to a role for MAP/microtubule affinity-regulating kinase 2 (MARK2/EMK/Par1b) in the regulation of metabolic events as well as in the control of CREB-dependent transcription activated by glucose in pancreatic islet beta cells. We have recently developed an in vitro kinase screening platform to identify novel kinase: Substrate pairs, the building blocks of signal transduction pathways. Application of this technology led us to identify MARK2 as the kinase that targets a novel glucose-regulated phosphorylation site on Transducer of Regulated CREB Activity 2 (TORC2, referred to as CREB-Regulated Transcriptional Coactivator 2, or CRTC2), a transcriptional coactivator essential for CREB activity in beta cells. We discuss these recent developments and suggest a model whereby members of the AMPK family integrate numerous signals to coordinate energy metabolism and cellular polarity with gene expression to regulate cell function/proliferation.


Journal of Biological Chemistry | 2016

High-throughput Functional Genomics Identifies Regulators of Primary Human Beta Cell Proliferation.

Karine Robitaille; Jillian L. Rourke; Joanne E. Mcbane; Accalia Fu; Stephen Baird; Qiujiang Du; Tatsuya Kin; A. M. James Shapiro; Robert A. Screaton

The expansion of cells for regenerative therapy will require the genetic dissection of complex regulatory mechanisms governing the proliferation of non-transformed human cells. Here, we report the development of a high-throughput RNAi screening strategy specifically for use in primary cells and demonstrate that silencing the cell cycle-dependent kinase inhibitors CDKN2C/p18 or CDKN1A/p21 facilitates cell cycle entry of quiescent adult human pancreatic beta cells. This work identifies p18 and p21 as novel targets for promoting proliferation of human beta cells and demonstrates the promise of functional genetic screens for dissecting therapeutically relevant state changes in primary human cells.


Diabetologia | 2015

LKB1 couples glucose metabolism to insulin secretion in mice.

Accalia Fu; Karine Robitaille; Brandon Faubert; Courtney Reeks; Xiao-Qing Dai; Alexandre B. Hardy; Krishana S. Sankar; Svetlana Ogrel; Osama Y. Al-Dirbashi; Jonathan V. Rocheleau; Michael B. Wheeler; Patrick E. MacDonald; Russell G. Jones; Robert A. Screaton

Aims/hypothesisPrecise regulation of insulin secretion by the pancreatic beta cell is essential for the maintenance of glucose homeostasis. Insulin secretory activity is initiated by the stepwise breakdown of ambient glucose to increase cellular ATP via glycolysis and mitochondrial respiration. Knockout of Lkb1, the gene encoding liver kinase B1 (LKB1) from the beta cell in mice enhances insulin secretory activity by an undefined mechanism. Here, we sought to determine the molecular basis for how deletion of Lkb1 promotes insulin secretion.MethodsTo explore the role of LKB1 on individual steps in the insulin secretion pathway, we used mitochondrial functional analyses, electrophysiology and metabolic tracing coupled with by gas chromatography and mass spectrometry.ResultsBeta cells lacking LKB1 surprisingly display impaired mitochondrial metabolism and lower ATP levels following glucose stimulation, yet compensate for this by upregulating both uptake and synthesis of glutamine, leading to increased production of citrate. Furthermore, under low glucose conditions, Lkb1−/− beta cells fail to inhibit acetyl-CoA carboxylase 1 (ACC1), the rate-limiting enzyme in lipid synthesis, and consequently accumulate NEFA and display increased membrane excitability.Conclusions/interpretationTaken together, our data show that LKB1 plays a critical role in coupling glucose metabolism to insulin secretion, and factors in addition to ATP act as coupling intermediates between feeding cues and secretion. Our data suggest that beta cells lacking LKB1 could be used as a system to identify additional molecular events that connect metabolism to cellular excitation in the insulin secretion pathway.


Cell Reports | 2015

Phospho-BAD BH3 Mimicry Protects β Cells and Restores Functional β Cell Mass in Diabetes

Sanda Ljubicic; Klaudia Polak; Accalia Fu; Jessica Wiwczar; Benjamin Szlyk; Yigang Chang; Juan C. Alvarez-Perez; Gregory H. Bird; Loren D. Walensky; Adolfo Garcia-Ocaña; Nika N. Danial

Strategies that simultaneously enhance the survival and glucose responsiveness of insulin-producing β cells will greatly augment β cell replacement therapies in type 1 diabetes (T1D). We show that genetic and pharmacologic mimetics of the phosphorylated BCL-2 homology 3 (BH3) domain of BAD impart β-cell-autonomous protective effects in the face of stress stimuli relevant to β cell demise in T1D. Importantly, these benefits translate into improved engraftment of donor islets in transplanted diabetic mice, increased β cell viability in islet grafts, restoration of insulin release, and diabetes reversal. Survival of β cells in this setting is not merely due to the inability of phospho-BAD to suppress prosurvival BCL-2 proteins but requires its activation of the glucose-metabolizing enzyme glucokinase. Thus, BAD phospho-BH3 mimetics may prove useful in the restoration of functional β cell mass in diabetes.


Cellular Signalling | 2011

Regulation of the CREB coactivator TORC by the dual leucine zipper kinase at different levels.

Do Thanh Phu; Manuel Wallbach; Chantal Depatie; Accalia Fu; Robert A. Screaton; Elke Oetjen

CREB is a ubiquitously expressed transcription factor regulating gene expression via binding to a CRE DNA element. Previous work showed that the dual leucine zipper kinase (DLK) reduced CREB-dependent gene transcription at least in part via inhibition of the coactivator CBP. Here we demonstrate that DLK also inhibits CREB activity by affecting the interaction of CREB with its second coactivator TORC. DLK acted on TORC-dependent transcription by distinct mechanisms. An interaction between DLK and all three TORC isoforms was demonstrated by in vitro protein-protein interaction assays and in cells by coimmunoprecipitation that required the N-terminus of TORC and the leucine zipper of dimerized DLK. Overexpressed DLK induced the phosphorylation of TORC2 and TORC1 on Ser-171 and 167, respectively and on additional residues. Since a kinase-dead DLK mutant did not prevent the nuclear localization of TORC and did not reduce TORC transcriptional activity to the same extent as wild-type DLK, we suggest that DLK-induced phosphorylation of TORC contributes to DLKs inhibitory action. Both the interaction with and the phosphorylation of TORC by DLK might account for the reduced recruitment of TORC to a CRE containing promoter as revealed by chromatin immunoprecipitation assay. These results show for the first time the inhibition of TORC function by a mitogen-activated kinase. Given the dependence on TORC in CREB-directed gene transcription, DLK and its downstream kinases thus contribute to the finely tuned regulation of CREB-dependent effects.

Collaboration


Dive into the Accalia Fu's collaboration.

Top Co-Authors

Avatar

Robert A. Screaton

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar

Chantal Depatie

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar

Courtney Reeks

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andy Cheuk-Him Ng

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar

Karine Robitaille

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar

Stephen Baird

Children's Hospital of Eastern Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chandra E. Eberhard

Children's Hospital of Eastern Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge