Network


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

Hotspot


Dive into the research topics where Feihan F. Dai is active.

Publication


Featured researches published by Feihan F. Dai.


Diabetologia | 2010

Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion

Nadeeja Wijesekara; Feihan F. Dai; Alexandre B. Hardy; P. R. Giglou; Alpana Bhattacharjee; V. Koshkin; Fabrice Chimienti; Herbert Y. Gaisano; Guy A. Rutter; Michael B. Wheeler

Aims/hypothesisZinc is highly concentrated in pancreatic beta cells, is critical for normal insulin storage and may regulate glucagon secretion from alpha cells. Zinc transport family member 8 (ZnT8) is a zinc efflux transporter that is highly abundant in beta cells. Polymorphisms of ZnT8 (also known as SLC30A8) gene in man are associated with increased risk of type 2 diabetes. While global Znt8 knockout (Znt8KO) mice have been characterised, ZnT8 is also present in other islet cell types and extra-pancreatic tissues. Therefore, it is important to find ways of understanding the role of ZnT8 in beta and alpha cells without the difficulties caused by the confounding effects of ZnT8 in these other tissues.MethodsWe generated mice with beta cell-specific (Znt8BKO) and alpha cell-specific (Znt8AKO) knockout of Znt8, and performed in vivo and in vitro characterisation of the phenotypes to determine the functional and anatomical impact of ZnT8 in these cells. Thus we assessed zinc accumulation, insulin granule morphology, insulin biosynthesis and secretion, and glucose homeostasis.ResultsZnt8BKO mice are glucose-intolerant, have reduced beta cell zinc accumulation and atypical insulin granules. They also display reduced first-phase glucose-stimulated insulin secretion, reduced insulin processing enzyme transcripts and increased proinsulin levels. In contrast, Znt8AKO mice show no evident abnormalities in plasma glucagon and glucose homeostasis.Conclusions/interpretationThis is the first report of specific beta and alpha cell deletion of Znt8. Our data indicate that while, under the conditions studied, ZnT8 is absolutely essential for proper beta cell function, it is largely dispensable for alpha cell function.


Journal of Biological Chemistry | 2008

Investigation of Transport Mechanisms and Regulation of Intracellular Zn2+ in Pancreatic α-Cells

Armen V. Gyulkhandanyan; Hongfang Lu; Simon Lee; Alpana Bhattacharjee; Nadeeja Wijesekara; Jocelyn E. Manning Fox; Patrick E. MacDonald; Fabrice Chimienti; Feihan F. Dai; Michael B. Wheeler

During insulin secretion, pancreatic α-cells are exposed to Zn2+ released from insulin-containing secretory granules. Although maintenance of Zn2+ homeostasis is critical for cell survival and glucagon secretion, very little is known about Zn2+-transporting pathways and the regulation of Zn2+ in α-cells. To examine the effect of Zn2+ on glucagon secretion and possible mechanisms controlling the intracellular Zn2+ level ([Zn2+]i), we employed a glucagon-producing cell line (α-TC6) and mouse islets where non-β-cells were identified using islets expressing green fluorescent protein exclusively in β-cells. In this study, we first confirmed that Zn2+ treatment resulted in the inhibition of glucagon secretion in α-TC6 cells and mouse islets in vitro. The inhibition of secretion was not likely via activation of KATP channels by Zn2+. We then determined that Zn2+ was transported into α-cells and was able to accumulate under both low and high glucose conditions, as well as upon depolarization of cells with KCl. The nonselective Ca2+ channel blocker Gd3+ partially inhibited Zn2+ influx in α-TC cells, whereas the L-type voltage-gated Ca2+ channel inhibitor nitrendipine failed to block Zn2+ accumulation. To investigate Zn2+ transport further, we profiled α-cells for Zn2+ transporter transcripts from the two families that work in opposite directions, SLC39 (ZIP, Zrt/Irt-like protein) and SLC30 (ZnT, Zn2+ transporter). We observed that Zip1, Zip10, and Zip14 were the most abundantly expressed Zips and ZnT4, ZnT5, and ZnT8 the dominant ZnTs. Because the redox state of cells is also a major regulator of [Zn2+]i, we examined the effects of oxidizing agents on Zn2+ mobilization within α-cells. 2,2′-Dithiodipyridine (-SH group oxidant), menadione (superoxide generator), and SIN-1 (3-morpholinosydnonimine) (peroxynitrite generator) all increased [Zn2+]i in α-cells. Together these results demonstrate that Zn2+ inhibits glucagon secretion, and it is transported into α-cells in part through Ca2+ channels. Zn2+ transporters and the redox state also modulate [Zn2+]i.


Cell Metabolism | 2014

The furan fatty acid metabolite CMPF is elevated in diabetes and induces β cell dysfunction.

Kacey J. Prentice; Lemieux Luu; Emma M. Allister; Ying Liu; Lucy S. Jun; Kyle W. Sloop; Alexandre B. Hardy; Li Wei; Weiping Jia; I. George Fantus; Douglas H. Sweet; Gary Sweeney; Ravi Retnakaran; Feihan F. Dai; Michael B. Wheeler

Gestational diabetes (GDM) results from failure of the β cells to adapt to increased metabolic demands; however, the cause of GDM and the extremely high rate of progression to type 2 diabetes (T2D) remains unknown. Using metabolomics, we show that the furan fatty acid metabolite 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) is elevated in the plasma of humans with GDM, as well as impaired glucose-tolerant and T2D patients. In mice, diabetic levels of plasma CMPF induced glucose intolerance, impaired glucose-stimulated insulin secretion, and decreased glucose utilization. Mechanistically, we show that CMPF acts directly on the β cell, causing impaired mitochondrial function, decreasing glucose-induced ATP accumulation, and inducing oxidative stress, resulting in dysregulation of key transcription factors and ultimately reduced insulin biosynthesis. Importantly, specifically blocking its transport through OAT3 or antioxidant treatment could prevent CMPF-induced β cell dysfunction. Thus, CMPF provides a link between β cell dysfunction and GDM/T2D that could be targeted therapeutically.


Journal of Biological Chemistry | 2008

Limited Mitochondrial Permeabilization Is an Early Manifestation of Palmitate-induced Lipotoxicity in Pancreatic β-Cells

Vasilij Koshkin; Feihan F. Dai; Christine A. Robson-Doucette; Catherine B. Chan; Michael B. Wheeler

Involvement of the mitochondrial permeability transition (MPT) pore in early stages of lipotoxic stress in the pancreatic β-cell lines MIN6 and INS-1 was the focus of this study. Both long term (indirect) and acute (direct) effects of fatty acid (FA) application on β-cell susceptibility to Ca2+-induced MPT induction were examined using both permeabilized and intact β-cells. Long term exposure to moderate (i.e. below cytotoxic) levels of the saturated FA palmitate sensitized β-cell mitochondria to MPT induced by Ca2+. Long term exposure to palmitate was significantly a more efficient inducer of MPT than the unsaturated FA oleate, although upon acute application both caused similar MPT activation. Application of antioxidants, inhibitors of the ceramide pathway, or modifiers of membrane fluidity did not protect β-cell mitochondria from FA exposure. However, significant protection was provided by co-application of the unsaturated FA oleate in a phosphatidylinositol 3-kinase-dependent manner. Characterization of MPT pore opening in response to moderate palmitate treatment revealed the opening of a unique form of MPT in β-cells as it encompassed features of both low and high conductance MPT states. Specifically, this MPT showed solute selectivity, characteristic of a low conductance MPT; however, it affected mitochondrial respiration and membrane potential in a way typical of a high conductance MPT. Activation of the full-size/high conductance form of MPT required application of high levels of FA that reduced growth and initiated apoptosis. These findings suggest that in the β-cell, MPTs can act as both initiators of cell death and as versatile modulators of cell metabolism, depending on the mode of the MPT pore induced.


Journal of Biological Chemistry | 2006

The Neuronal Ca2+ Sensor Protein Visinin-like Protein-1 Is Expressed in Pancreatic Islets and Regulates Insulin Secretion

Feihan F. Dai; Yi Zhang; Youhou Kang; Qinghua Wang; Herbert Y. Gaisano; Karl-Heinz Braunewell; Catherine B. Chan; Michael B. Wheeler

Visinin-like protein-1 (VILIP-1) is a member of the neuronal Ca2+ sensor protein family that modulates Ca2+-dependent cell signaling events. VILIP-1, which is expressed primarily in the brain, increases cAMP formation in neural cells by modulating adenylyl cyclase, but its functional role in other tissues remains largely unknown. In this study, we demonstrate that VILIP-1 is expressed in murine pancreatic islets and β-cells. To gain insight into the functions of VILIP-1 in β-cells, we used both overexpression and small interfering RNA knockdown strategies. Overexpression of VILIP-1 in the MIN6β-cell line or isolated mouse islets had no effect on basal insulin secretion but significantly increased glucose-stimulated insulin secretion. cAMP accumulation was elevated in VILIP-1-overexpressing cells, and the protein kinase A inhibitor H-89 attenuated increased glucose-stimulated insulin secretion. Overexpression of VILIP-1 in isolated mouse β-cells increased cAMP content accompanied by increased cAMP-responsive element-binding protein gene expression and enhanced exocytosis as detected by cell capacitance measurements. Conversely, VILIP-1 knockdown by small interfering RNA caused a reduction in cAMP accumulation and produced a dramatic increase in preproinsulin mRNA, basal insulin secretion, and total cellular insulin content. The increase in preproinsulin mRNA in these cells was attributed to enhanced insulin gene transcription. Taken together, we have shown that VILIP-1 is expressed in pancreatic β-cells and modulates insulin secretion. Increased VILIP-1 enhanced insulin secretion in a cAMP-associated manner. Down-regulation of VILIP-1 was accompanied by decreased cAMP accumulation but increased insulin gene transcription.


Diabetologia | 2013

The loss of Sirt1 in mouse pancreatic beta cells impairs insulin secretion by disrupting glucose sensing

Lemieux Luu; Feihan F. Dai; Kacey J. Prentice; X. Huang; Alexandre B. Hardy; Jakob Bondo Hansen; Ying Liu; Jamie W. Joseph; Michael B. Wheeler

Aims/hypothesisSirtuin 1 (SIRT1) has emerged as a key metabolic regulator of glucose homeostasis and insulin secretion. Enhanced SIRT1 activity has been shown to be protective against diabetes, although the mechanisms remain largely unknown. The aim of this study was to determine how SIRT1 regulates insulin secretion in the pancreatic beta cell.MethodsPancreatic beta cell-specific Sirt1 deletion was induced by tamoxifen injection in 9-week-old Pdx1CreER:floxSirt1 mice (Sirt1BKO). Controls were injected with vehicle. Mice were assessed metabolically via glucose challenge, insulin tolerance tests and physical variables. In parallel, Sirt1 short interfering RNA-treated MIN6 cells (SIRT1KD) and isolated Sirt1BKO islets were used to investigate the effect of SIRT1 inactivation on insulin secretion and gene expression.ResultsOGTTs showed impaired glucose disposal in Sirt1BKO mice due to insufficient insulin secretion. Isolated Sirt1BKO islets and SIRT1KD MIN6 cells also exhibited impaired glucose-stimulated insulin secretion. Subsequent analyses revealed impaired α-ketoisocaproic acid-induced insulin secretion and attenuated glucose-induced Ca2+ influx, but normal insulin granule exocytosis in Sirt1BKO beta cells. Microarray studies revealed a large cluster of mitochondria-related genes, the expression of which was dysregulated in SIRT1KD MIN6 cells. Upon further analysis, we demonstrated an explicit defect in mitochondrial function: the inability to couple nutrient metabolism to mitochondrial membrane hyperpolarisation and reduced oxygen consumption rates.Conclusions/interpretationTaken together, these findings indicate that in beta cells the deacetylase SIRT1 regulates the expression of specific mitochondria-related genes that control metabolic coupling, and that a decrease in beta cell Sirt1 expression impairs glucose sensing and insulin secretion.


Journal of Biological Chemistry | 2015

Characterization of Zinc Influx Transporters (ZIPs) in Pancreatic β Cells: ROLES IN REGULATING CYTOSOLIC ZINC HOMEOSTASIS AND INSULIN SECRETION.

Ying Liu; Battsetseg Batchuluun; Louisa Ho; Dan Zhu; Kacey J. Prentice; Alpana Bhattacharjee; Ming Zhang; Farzaneh Pourasgari; Alexandre B. Hardy; Kathryn Mary Taylor; Herbert Y. Gaisano; Feihan F. Dai; Michael B. Wheeler

Background: Zinc influx transporters (ZIPs), together with zinc efflux transporters (ZnTs), regulate cellular zinc homeostasis. Results: Down-regulation of ZIP6 and ZIP7 expression impairs glucose-stimulated insulin secretion via reduced first-phase insulin exocytosis. Conclusion: ZIP6 and ZIP7 are functionally important for maintaining proper insulin secretion in pancreatic β cells. Significance: ZIP6 and ZIP7 represent novel proteins that contribute to the insulin secretory pathway. Zinc plays an essential role in the regulation of pancreatic β cell function, affecting important processes including insulin biosynthesis, glucose-stimulated insulin secretion, and cell viability. Mutations in the zinc efflux transport protein ZnT8 have been linked with both type 1 and type 2 diabetes, further supporting an important role for zinc in glucose homeostasis. However, very little is known about how cytosolic zinc is controlled by zinc influx transporters (ZIPs). In this study, we examined the β cell and islet ZIP transcriptome and show consistent high expression of ZIP6 (Slc39a6) and ZIP7 (Slc39a7) genes across human and mouse islets and MIN6 β cells. Modulation of ZIP6 and ZIP7 expression significantly altered cytosolic zinc influx in pancreatic β cells, indicating an important role for ZIP6 and ZIP7 in regulating cellular zinc homeostasis. Functionally, this dysregulated cytosolic zinc homeostasis led to impaired insulin secretion. In parallel studies, we identified both ZIP6 and ZIP7 as potential interacting proteins with GLP-1R by a membrane yeast two-hybrid assay. Knock-down of ZIP6 but not ZIP7 in MIN6 β cells impaired the protective effects of GLP-1 on fatty acid-induced cell apoptosis, possibly via reduced activation of the p-ERK pathway. Therefore, our data suggest that ZIP6 and ZIP7 function as two important zinc influx transporters to regulate cytosolic zinc concentrations and insulin secretion in β cells. In particular, ZIP6 is also capable of directly interacting with GLP-1R to facilitate the protective effect of GLP-1 on β cell survival.


Diabetes | 2016

A Predictive Metabolic Signature for the Transition From Gestational Diabetes Mellitus to Type 2 Diabetes.

Amina Allalou; Amarnadh Nalla; Kacey J. Prentice; Ying Liu; Ming Zhang; Feihan F. Dai; Xian Ning; Lucy R. Osborne; Brian J. Cox; Erica P. Gunderson; Michael B. Wheeler

Gestational diabetes mellitus (GDM) affects 3–14% of pregnancies, with 20–50% of these women progressing to type 2 diabetes (T2D) within 5 years. This study sought to develop a metabolomics signature to predict the transition from GDM to T2D. A prospective cohort of 1,035 women with GDM pregnancy were enrolled at 6–9 weeks postpartum (baseline) and were screened for T2D annually for 2 years. Of 1,010 women without T2D at baseline, 113 progressed to T2D within 2 years. T2D developed in another 17 women between 2 and 4 years. A nested case-control design used 122 incident case patients matched to non–case patients by age, prepregnancy BMI, and race/ethnicity. We conducted metabolomics with baseline fasting plasma and identified 21 metabolites that significantly differed by incident T2D status. Machine learning optimization resulted in a decision tree modeling that predicted T2D incidence with a discriminative power of 83.0% in the training set and 76.9% in an independent testing set, which is far superior to measuring fasting plasma glucose levels alone. The American Diabetes Association recommends T2D screening in the early postpartum period via oral glucose tolerance testing after GDM, which is a time-consuming and inconvenient procedure. Our metabolomics signature predicted T2D incidence from a single fasting blood sample. This study represents the first metabolomics study of the transition from GDM to T2D validated in an independent testing set, facilitating early interventions.


Cell Reports | 2016

Rapid Elevation in CMPF May Act As a Tipping Point in Diabetes Development.

Ying Liu; Kacey J. Prentice; Judith A. Eversley; Cheng Hu; Battsetseg Batchuluun; Katherine Leavey; Jakob Bondo Hansen; David Wei; Brian J. Cox; Feihan F. Dai; Weiping Jia; Michael B. Wheeler

Prediabetes, a state of mild glucose intolerance, can persist for years before a sudden decline in beta cell function and rapid deterioration to overt diabetes. The mechanism underlying this tipping point of beta cell dysfunction remains unknown. Here, the furan fatty acid metabolite CMPF was evaluated in a prospective cohort. Those who developed overt diabetes had a significant increase in CMPF over time, whereas prediabetics maintained chronically elevated levels, even up to 5 years before diagnosis. To evaluate the effect of increasing CMPF on diabetes progression, we used obese, insulin-resistant models of prediabetes. CMPF accelerated diabetes development by inducing metabolic remodeling, resulting in preferential utilization of fatty acids over glucose. This was associated with diminished glucose-stimulated insulin secretion, increased ROS formation, and accumulation of proinsulin, all characteristics of human diabetes. Thus, an increase in CMPF may represent the tipping point in diabetes development by accelerating beta cell dysfunction.


Molecular Endocrinology | 2013

The identification of novel proteins that interact with the GLP-1 receptor and restrain its activity.

X. Huang; Feihan F. Dai; G. Gaisano; K. Giglou; J. Han; Ming Zhang; S. Kittanakom; V. Wong; Li Wei; Aaron D. Showalter; Kyle W. Sloop; I. Stagljar; Michael B. Wheeler

Glucagon-like peptide 1 receptor (GLP-1R) controls diverse physiological functions in tissues including the pancreatic islets, brain, and heart. To understand the mechanisms that control glucagon-like peptide 1 (GLP-1) signaling better, we sought to identify proteins that interact with the GLP-1R using a membrane-based split ubiquitin yeast two-hybrid (MYTH) assay. A screen of a human fetal brain cDNA prey library with an unliganded human GLP-1R as bait in yeast revealed 38 novel interactor protein candidates. These interactions were confirmed in mammalian Chinese hamster ovarian cells by coimmunoprecipitation. Immunofluorescence was used to show subcellular colocalization of the interactors with GLP-1R. Cluster analysis revealed that the interactors were primarily associated with signal transduction, metabolism, and cell development. When coexpressed with the GLP-1R in Chinese hamster ovarian cells, 15 interactors significantly altered GLP-1-induced cAMP accumulation. Surprisingly, all 15 proteins inhibited GLP-1-activated cAMP. Given GLP-1s prominent role as an incretin, we then focused on 3 novel interactors, SLC15A4, APLP1, and AP2M1, because they are highly expressed and localized to the membrane in mouse insulinoma β-cells. Small interfering RNA-mediated knockdown of each candidate gene significantly enhanced GLP-1-induced insulin secretion. In conclusion, we have generated a novel GLP-1R-protein interactome, identifying several interactors that suppress GLP-1R signaling. We suggest that the inhibition of these interactors may serve as a novel strategy to enhance GLP-1R activity.

Collaboration


Dive into the Feihan F. Dai's collaboration.

Top Co-Authors

Avatar

Michael B. Wheeler

University of Prince Edward Island

View shared research outputs
Top Co-Authors

Avatar

Ying Liu

University of Toronto

View shared research outputs
Top Co-Authors

Avatar

Alpana Bhattacharjee

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

X. Huang

University of Toronto

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Wei

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge