Network


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

Hotspot


Dive into the research topics where Arturo Mancini is active.

Publication


Featured researches published by Arturo Mancini.


Trends in Endocrinology and Metabolism | 2013

The fatty acid receptor FFA1/GPR40 a decade later: how much do we know?

Arturo Mancini; Vincent Poitout

Glucose homeostasis requires the highly coordinated regulation of insulin secretion by pancreatic β cells. This is primarily mediated by glucose itself, but other nutrients, including free fatty acids (FFAs), potentiate the insulinotropic capacity of glucose. A decade ago, the seven-transmembrane domain receptor (7TMR) GPR40 was demonstrated to be predominantly expressed in β cells and activated by long-chain FFAs. This discovery added a new dimension to our understanding of FFA-mediated control of glucose homeostasis. Furthermore, GPR40 has drawn considerable interest as a novel therapeutic target to enhance insulin secretion in type 2 diabetes. However, our understanding of the biology of GPR40 remains incomplete and its physiological role controversial. Here we summarize the current state of knowledge and emerging concepts regarding the role of GPR40 in regulating glucose homeostasis.


Journal of Biological Chemistry | 2015

β-Arrestin Recruitment and Biased Agonism at Free Fatty Acid Receptor 1

Arturo Mancini; Gyslaine Bertrand; Kevin Vivot; Eric Carpentier; Caroline Tremblay; Julien Ghislain; Michel Bouvier; Vincent Poitout

Background: FFAR1/GPR40 is a potential target to enhance insulin secretion in type 2 diabetes, yet knowledge of the pharmacobiology of GPR40 remains incomplete. Results: GPR40 functions via both G protein-mediated and β-arrestin-mediated mechanisms; endogenous and synthetic ligands differentially engage these pathways to promote insulin secretion. Conclusion: GPR40 is subject to functionally relevant biased agonism. Significance: Biased agonism at GPR40 could be exploited for therapeutic purposes. FFAR1/GPR40 is a seven-transmembrane domain receptor (7TMR) expressed in pancreatic β cells and activated by FFAs. Pharmacological activation of GPR40 is a strategy under consideration to increase insulin secretion in type 2 diabetes. GPR40 is known to signal predominantly via the heterotrimeric G proteins Gq/11. However, 7TMRs can also activate functionally distinct G protein-independent signaling via β-arrestins. Further, G protein- and β-arrestin-based signaling can be differentially modulated by different ligands, thus eliciting ligand-specific responses (“biased agonism”). Whether GPR40 engages β-arrestin-dependent mechanisms and is subject to biased agonism is unknown. Using bioluminescence resonance energy transfer-based biosensors for real-time monitoring of cell signaling in living cells, we detected a ligand-induced GPR40-β-arrestin interaction, with the synthetic GPR40 agonist TAK-875 being more effective than palmitate or oleate in recruiting β-arrestins 1 and 2. Conversely, TAK-875 acted as a partial agonist of Gq/11-dependent GPR40 signaling relative to both FFAs. Pharmacological blockade of Gq activity decreased FFA-induced insulin secretion. In contrast, knockdown or genetic ablation of β-arrestin 2 in an insulin-secreting cell line and mouse pancreatic islets, respectively, uniquely attenuated the insulinotropic activity of TAK-875, thus providing functional validation of the biosensor data. Collectively, these data reveal that in addition to coupling to Gq/11, GPR40 is functionally linked to a β-arrestin 2-mediated insulinotropic signaling axis. These observations expose previously unrecognized complexity for GPR40 signal transduction and may guide the development of biased agonists showing improved clinical profile in type 2 diabetes.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Glucose activates free fatty acid receptor 1 gene transcription via phosphatidylinositol-3-kinase-dependent O-GlcNAcylation of pancreas-duodenum homeobox-1

Melkam Kebede; Mourad Ferdaoussi; Arturo Mancini; Thierry Alquier; Rohit N. Kulkarni; Michael D. Walker; Vincent Poitout

The G protein-coupled free fatty acid receptor-1 (FFA1/GPR40) plays a major role in the regulation of insulin secretion by fatty acids. GPR40 is considered a potential therapeutic target to enhance insulin secretion in type 2 diabetes; however, its mode of regulation is essentially unknown. The aims of this study were to test the hypothesis that glucose regulates GPR40 gene expression in pancreatic β-cells and to determine the mechanisms of this regulation. We observed that glucose stimulates GPR40 gene transcription in pancreatic β-cells via increased binding of pancreas-duodenum homeobox-1 (Pdx-1) to the A-box in the HR2 region of the GPR40 promoter. Mutation of the Pdx-1 binding site within the HR2 abolishes glucose activation of GPR40 promoter activity. The stimulation of GPR40 expression and Pdx-1 binding to the HR2 in response to glucose are mimicked by N-acetyl glucosamine, an intermediate of the hexosamine biosynthesis pathway, and involve PI3K-dependent O-GlcNAcylation of Pdx-1 in the nucleus. We demonstrate that O-GlcNAc transferase (OGT) interacts with the product of the PI3K reaction, phosphatidylinositol 3,4,5-trisphosphate (PIP3), in the nucleus. This interaction enables OGT to catalyze O-GlcNAcylation of nuclear proteins, including Pdx-1. We conclude that glucose stimulates GPR40 gene expression at the transcriptional level through Pdx-1 binding to the HR2 region and via a signaling cascade that involves an interaction between OGT and PIP3 at the nuclear membrane. These observations reveal a unique mechanism by which glucose metabolism regulates the function of transcription factors in the nucleus to induce gene expression.


Journal of Biological Chemistry | 2010

Leukotriene B4 BLT Receptor Signaling Regulates the Level and Stability of Cyclooxygenase-2 (COX-2) mRNA through Restricted Activation of Ras/Raf/ERK/p42 AUF1 Pathway

Beibei Zhai; Huiqing Yang; Arturo Mancini; QingWen He; John Antoniou; John A. Di Battista

Recent studies suggest that active resolution of the inflammatory response in animal models of arthritis may involve leukotriene B4 (LTB4)-dependent stimulation of “intermediate” prostaglandin production, which in turn favors the synthesis of “downstream” anti-inflammatory and pro-resolving lipoxins, resolvins, and protectins. We explored a putative mechanism involving LTB4-dependent control of cyclooxygenase-2 (COX-2) expression, the rate-limiting step in inflammatory prostaglandin biosynthesis. Indeed, LTB4 potently up-regulated/stabilized interleukin-1β-induced COX-2 mRNA and protein expression under conditions of COX-2 inhibitor-dependent blockade of PGE2 release in human synovial fibroblasts (EC50 = 16.5 ± 1.7 nm for mRNA; 19 ± 2.4 nm for protein, n = 4). The latter response was pertussis toxin-sensitive, and semi-quantitative reverse transcription-PCR confirmed the quantitative predominance of the BLT2 receptor. Transfection experiments, using human COX-2 promoter plasmids and chimeric luciferase-COX-2 mRNA 3′-untranslated region (3′-UTR) reporter constructs, revealed that LTB4 exerted its stabilizing effect at the post-transcriptional level through a 116-bp adenylate/uridylate-rich sequence in the proximal region of the COX-2 3′-UTR. Using luciferase-COX-2 mRNA 3′-UTR reporter constructs and Ras/c-Raf expression and mutant constructs, we showed that the Ras/c-Raf/MEK1/2/ERK1/2 signaling pathway mediated LTB4-dependent COX-2 mRNA stabilization. Knockdown experiments with specific short hairpin RNAs confirmed that LTB4 stabilization of COX-2 mRNA was apparently mediated through the RNA-binding protein, p42 AUF1. The nuclear export of p42 AUF1 was driven by c-Raf/MEK1/2/ERK1/2 signaling and sensitive to leptomycin B treatment, suggesting a CRM1-dependent mechanism. We conclude that LTB4 may support the resolution phase of the inflammatory response by stabilizing COX-2, ensuring a reservoir of ambient pro-resolution lipid mediators.


Journal of Biological Chemistry | 2006

Prostaglandin E2 Stimulates p53 Transactivational Activity through Specific Serine 15 Phosphorylation in Human Synovial Fibroblasts ROLE IN SUPPRESSION OF c/EBP/NF-κB-MEDIATED MEKK1-INDUCED MMP-1 EXPRESSION

Wissam H. Faour; QingWen He; Arturo Mancini; Dragan Jovanovic; John Antoniou; John A. Di Battista

Cyclooxygenase-2 (COX-2) overexpression has been linked to cell survival, transformation, and hyperproliferation. We examined the regulation of the tumor suppressor gene p53 and p53 target genes by prostaglandin E2 (PGE2) in human synovial fibroblasts (HSF). PGE2 induced a time-dependent increase in p53 Ser15 phosphorylation, with no discernible change in overall p53 levels. PGE2-dependent Ser15 phosphorylation was apparently mediated by activated p38 MAP kinase as SB202190, a p38 kinase inhibitor, blocked the response. Overexpression of a MKK3 construct, but not MKK1, stimulated SB202190-sensitive p53 Ser15 phosphorylation. PGE2-stimulated [phospho-Ser15]p53 transactivated a p53 response element (GADD45)-luciferase reporter in transiently transfected HSF (SN7); the effect was compromised by overexpression of a dominant-negative mutant (dnm) of p53 or excess p53S15A expression plasmid but mimicked by a constitutively active p53S15E expression construct. PGE2, wtp53 expression in the presence of PGE2, and p53S15E suppressed steady-state levels of MEKK1-induced MMP-1 mRNA, effects nullified with co-transfection of p53 dnm or p53S15A. MEKK1-induced MMP-1 promoter-driven luciferase activity was largely dependent on a c/EBPβ-NF-κB-like enhancer site at –2008 to –1972 bp, as judged by deletion and point mutation analyses. PGE2, overexpression of p53wt with PGE2, or p53S15E abolished the MEKK1-induced MMP-1 promoter luciferase activity. Gel-shift/super gel-shift analyses identified c/EBPβ dimers and c/EBPβ/NF-κB p65 heterodimers as binding species at the apparent site of MEKK1-dependent transactivation. PGE2-stimulated [phospho-Ser15]p53 abrogated the DNA binding of c/EBPβ dimers and c/EBPβ/NF-κB p65 heterodimers. Our data suggest that COX-2 prostaglandins may be implicated in p53 function and p53 target gene expression.


The Journal of Rheumatology | 2011

CD101 expression and function in normal and rheumatoid arthritis-affected human T cells and monocytes/macrophages.

Dragan Jovanovic; Laurence Boumsell; Armand Bensussan; Xavier Chevalier; Arturo Mancini; John A. Di Battista

Objective. It was recently reported that CD101 surface expression discriminates potency among CD4+CD25+ FoxP3+ regulatory T cells in the mouse. We investigated whether CD101 may also have a role in the suppressor function of regulatory T cells in humans given that the latter population may affect the autoimmune response in patients with rheumatoid arthritis (RA). Methods. Sorted T cells and monocyte/macrophage cell populations were analyzed by flow cyto metry using conjugated antibodies specific for cell-surface markers. T cell proliferation assays were conducted by [3H]thymidine incorporation and CD8high cytotoxicity measurements by Cyto-Scan-LDH cytotoxicity assays. ELISA were used to measure cytokines in cell culture supernatants and Western blotting was performed for profiling mitogen-activated protein (MAP) kinase activation using specific antiphospholipid antibodies. Results. CD101 expression coincided with PMA-induced monocyte/leukocyte lineage differentiation. CD8highCD101− T cells exhibited greater cytotoxic activity than CD8highCD101+ T cells, while no difference was observed between CD4CD25highCD101+ and CD4CD25highCD101− Treg inhibitory activity through responder T cells. LPS-induced proinflammatory cytokine production and p38 MAP kinase activation were made possible by ligation of CD101 with an anti-CD101 antibody F(ab’)2 fragment. Conclusion. These results suggested a modulatory/coregulatory function of CD101 in the human immune system, in contrast to murine models, in which CD101 surface expression discriminates potency among FoxP3+ regulatory T cells. Cytotoxic CD8highCD101+ T cells were markedly less cytotoxic than CD8high T cells negative for the CD101 antigen and were conspicuously downregulated in patients with RA, suggesting a possible role for CD101 expression and function in the control of certain manifestations of RA pathology.


Molecular metabolism | 2016

The regulator of G-protein signaling RGS16 promotes insulin secretion and β-cell proliferation in rodent and human islets

Kevin Vivot; Valentine S. Moullé; Bader Zarrouki; Caroline Tremblay; Arturo Mancini; Hasna Maachi; Julien Ghislain; Vincent Poitout

Objective G protein-coupled receptor (GPCR) signaling regulates insulin secretion and pancreatic β cell-proliferation. While much knowledge has been gained regarding how GPCRs are activated in β cells, less is known about the mechanisms controlling their deactivation. In many cell types, termination of GPCR signaling is controlled by the family of Regulators of G-protein Signaling (RGS). RGS proteins are expressed in most eukaryotic cells and ensure a timely return to the GPCR inactive state upon removal of the stimulus. The aims of this study were i) to determine if RGS16, the most highly enriched RGS protein in β cells, regulates insulin secretion and β-cell proliferation and, if so, ii) to elucidate the mechanisms underlying such effects. Methods Mouse and human islets were infected with recombinant adenoviruses expressing shRNA or cDNA sequences to knock-down or overexpress RGS16, respectively. 60 h post-infection, insulin secretion and cAMP levels were measured in static incubations in the presence of glucose and various secretagogues. β-cell proliferation was measured in infected islets after 72 h in the presence of 16.7 mM glucose ± somatostatin and various inhibitors. Results RGS16 mRNA levels are strongly up-regulated in islets of Langerhans under hyperglycemic conditions in vivo and ex vivo. RGS16 overexpression stimulated glucose-induced insulin secretion in isolated mouse and human islets while, conversely, insulin secretion was impaired following RGS16 knock-down. Insulin secretion was no longer affected by RGS16 knock-down when islets were pre-treated with pertussis toxin to inactivate Gαi/o proteins, or in the presence of a somatostatin receptor antagonist. RGS16 overexpression increased intracellular cAMP levels, and its effects were blocked by an adenylyl cyclase inhibitor. Finally, RGS16 overexpression prevented the inhibitory effect of somatostatin on insulin secretion and β-cell proliferation. Conclusions Our results identify RGS16 as a novel regulator of β-cell function that coordinately controls insulin secretion and proliferation by limiting the tonic inhibitory signal exerted by δ-cell-derived somatostatin in islets.


Journal of Biological Chemistry | 2003

T-cell-derived Interleukin-17 Regulates the Level and Stability of Cyclooxygenase-2 (COX-2) mRNA through Restricted Activation of the p38 Mitogen-activated Protein Kinase Cascade ROLE OF DISTAL SEQUENCES IN THE 3′-UNTRANSLATED REGION OF COX-2 mRNA

Wissam H. Faour; Arturo Mancini; Qing Wen He; John A. Di Battista


Journal of Biological Chemistry | 2005

Early Growth Response Factor-1 Mediates Prostaglandin E2-dependent Transcriptional Suppression of Cytokine-induced Tumor Necrosis Factor-α Gene Expression in Human Macrophages and Rheumatoid Arthritis-affected Synovial Fibroblasts

Wissam H. Faour; Nada Alaaeddine; Arturo Mancini; Qing Wen He; Dragan Jovanovic; John A. Di Battista


American Journal of Physiology-renal Physiology | 2007

The 3′-untranslated region of the Ste20-like kinase SLK regulates SLK expression

Andrey V. Cybulsky; Tomoko Takano; Joan Papillon; Wen Hao; Arturo Mancini; John A. Di Battista; Myron I. Cybulsky

Collaboration


Dive into the Arturo Mancini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin Vivot

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Michel Bouvier

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beibei Zhai

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge