Network


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

Hotspot


Dive into the research topics where Emilyn U. Alejandro is active.

Publication


Featured researches published by Emilyn U. Alejandro.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Insulin protects islets from apoptosis via Pdx1 and specific changes in the human islet proteome

James D. Johnson; Ernesto Bernal-Mizrachi; Emilyn U. Alejandro; Zhiqiang Han; Tatyana B. Kalynyak; Hong Li; Jennifer L. Beith; Julia Gross; Garth L. Warnock; R. Reid Townsend; M. Alan Permutt; Kenneth S. Polonsky

Insulin is both a hormone regulating energy metabolism and a growth factor. We and others have shown that physiological doses of insulin initiate complex signals in primary human and mouse β-cells, but the functional significance of insulins effects on this cell type remains unclear. In the present study, the role of insulin in β-cell apoptosis was examined. Exogenous insulin completely prevented apoptosis induced by serum withdrawal when given at picomolar or low nanomolar concentrations but not at higher concentrations, indicating that physiological concentrations of insulin are antiapoptotic and that insulin signaling is self-limiting in islets. Insulin treatment was associated with the nuclear localization of Pdx1 and the prosurvival effects of insulin were largely absent in islets 50% deficient in Pdx1, providing direct evidence that Pdx1 is a signaling target of insulin. Physiological levels of insulin did not increase Akt phosphorylation, and the protective effects of insulin were only partially altered in islets lacking 80% of normal Akt activity, suggesting the presence of additional insulin-regulated antiapoptotic pathways. Proteomic analysis of insulin-treated human islets revealed significant changes in multiple proteins. Bridge-1, a Pdx1-binding partner and regulator of β-cell survival, was increased significantly at low insulin doses. Together, these data suggest that insulin can act as a master regulator of islet survival by regulating Pdx1.


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

Carboxypeptidase E mediates palmitate-induced β-cell ER stress and apoptosis

Kristin D. Jeffrey; Emilyn U. Alejandro; Dan S. Luciani; Tatyana B. Kalynyak; Xiaoke Hu; Hong Li; Yalin Lin; R. Reid Townsend; Kenneth S. Polonsky; James D. Johnson

Obesity is a principal risk factor for type 2 diabetes, and elevated fatty acids reduce β-cell function and survival. An unbiased proteomic screen was used to identify targets of palmitate in β-cell death. The most significantly altered protein in both human islets and MIN6 β-cells treated with palmitate was carboxypeptidase E (CPE). Palmitate reduced CPE protein levels within 2 h, preceding endoplasmic reticulum (ER) stress and cell death, by a mechanism involving CPE translocation to Golgi and lysosomal degradation. Palmitate metabolism and Ca2+ flux were also required for CPE proteolysis and β-cell death. Chronic palmitate exposure increased the ratio of proinsulin to insulin. CPE null islets had increased apoptosis in vivo and in vitro. Reducing CPE by ≈30% using shRNA also increased ER stress and apoptosis. Conversely, overexpression of CPE partially rescued β-cells from palmitate-induced ER stress and apoptosis. Thus, carboxypeptidase E degradation contributes to palmitate-induced β-cell ER stress and apoptosis. CPE is a major link between hyperlipidemia and β-cell death pathways in diabetes.


Endocrinology | 2008

Insulin Stimulates Primary β-Cell Proliferation via Raf-1 Kinase

Jennifer L. Beith; Emilyn U. Alejandro; James D. Johnson

A relative decrease in beta-cell mass is key in the pathogenesis of type 1 diabetes, type 2 diabetes, and in the failure of transplanted islet grafts. It is now clear that beta-cell duplication plays a dominant role in the regulation of adult beta-cell mass. Therefore, knowledge of the endogenous regulators of beta-cell replication is critical for understanding the physiological control of beta-cell mass and for harnessing this process therapeutically. We have shown that concentrations of insulin known to exist in vivo act directly on beta-cells to promote survival. Whether insulin stimulates adult beta-cell proliferation remains unclear. We tested this hypothesis using dispersed primary mouse islet cells double labeled with 5-bromo-2-deoxyuridine and insulin antisera. Treating cells with 200-pm insulin significantly increased proliferation from a baseline rate of 0.15% per day. Elevating glucose from 5-15 mm did not significantly increase beta-cell replication. beta-Cell proliferation was inhibited by somatostatin as well as inhibitors of insulin signaling. Interestingly, inhibiting Raf-1 kinase blocked proliferation stimulated by low, but not high (superphysiological), insulin doses. Insulin-stimulated mouse insulinoma cell proliferation was dependent on both phosphatidylinositol 3-kinase/Akt and Raf-1/MAPK kinase pathways. Overexpression of Raf-1 was sufficient to increase proliferation in the absence of insulin, whereas a dominant-negative Raf-1 reduced proliferation in the presence of 200-pm insulin. Together, these results demonstrate for the first time that insulin, at levels that have been measured in vivo, can directly stimulate beta-cell proliferation and that Raf-1 kinase is involved in this process. These findings have significant implications for the understanding of the regulation of beta-cell mass in both the hyperinsulinemic and insulin-deficient states that occur in the various forms of diabetes.


Molecular Aspects of Medicine | 2015

Natural history of β-cell adaptation and failure in type 2 diabetes

Emilyn U. Alejandro; Brigid Gregg; Manuel Blandino-Rosano; Corentin Cras-Méneur; Ernesto Bernal-Mizrachi

Type 2 diabetes mellitus (T2D) is a complex disease characterized by β-cell failure in the setting of insulin resistance. The current evidence suggests that genetic predisposition, and environmental factors can impair the capacity of the β-cells to respond to insulin resistance and ultimately lead to their failure. However, genetic studies have demonstrated that known variants account for less than 10% of the overall estimated T2D risk, suggesting that additional unidentified factors contribute to susceptibility of this disease. In this review, we will discuss the different stages that contribute to the development of β-cell failure in T2D. We divide the natural history of this process in three major stages: susceptibility, β-cell adaptation and β-cell failure, and provide an overview of the molecular mechanisms involved. Further research into mechanisms will reveal key modulators of β-cell failure and thus identify possible novel therapeutic targets and potential interventions to protect against β-cell failure.


Journal of Biological Chemistry | 2008

Inhibition of Raf-1 Alters Multiple Downstream Pathways to Induce Pancreatic β-Cell Apoptosis

Emilyn U. Alejandro; James D. Johnson

The serine threonine kinase Raf-1 plays a protective role in many cell types, but its function in pancreatic β-cells has not been elucidated. In the present study, we examined whether primary β-cells possess Raf-1 and tested the hypothesis that Raf-1 is critical for β-cell survival. Using reverse transcriptase-PCR, Western blot, and immunofluorescence, we identified Raf-1 in human islets, mouse islets, and in the MIN6 β-cell line. Blocking Raf-1 activity using a specific Raf-1 inhibitor or dominant-negative Raf-1 mutants led to a time- and dose-dependent increase in cell death, assessed by real-time imaging of propidium iodide incorporation, TUNEL, PCR-enhanced DNA laddering, and Caspase-3 cleavage. Although the rapid increase in apoptotic cell death was associated with decreased Erk phosphorylation, studies with two Mek inhibitors suggested that the classical Erk-dependent pathway could explain only part of the cell death observed after inhibition of Raf-1. An alternative Erk-independent pathway downstream of Raf-1 kinase involving the pro-apoptotic protein Bad has recently been characterized in other tissues. Inhibiting Raf-1 in β-cells led to a striking loss of Bad phosphorylation at serine 112 and an increase in the protein levels of both Bad and Bax. Together, our data strongly suggest that Raf-1 signaling plays an important role regulating β-cell survival, via both Erk-dependent and Bad-dependent mechanisms. Conversely, acutely inhibiting phosphatidylinositol 3-kinase Akt had more modest effects on β-cell death. These studies identify Raf-1 as a critical anti-apoptotic kinase in pancreatic β-cells and contribute to our understanding of survival signaling in this cell type.


Cell Cycle | 2012

mTORC1 signaling and regulation of pancreatic β-cell mass

Manuel Blandino-Rosano; Angela Y. Chen; Joshua O. Scheys; Emilyn U. Alejandro; Aaron Gould; Tatyana Taranukha; Lynda Elghazi; Corentin Cras-Méneur; Ernesto Bernal-Mizrachi

The capacity of β cells to expand in response to insulin resistance is a critical factor in the development of type 2 diabetes. Proliferation of β cells is a major component for these adaptive responses in animal models. The extracellular signals responsible for β-cell expansion include growth factors, such as insulin, and nutrients, such as glucose and amino acids. AKT activation is one of the important components linking growth signals to the regulation of β-cell expansion. Downstream of AKT, tuberous sclerosis complex 1 and 2 (TSC1/2) and mechanistic target of rapamycin complex 1 (mTORC1) signaling have emerged as prime candidates in this process, because they integrate signals from growth factors and nutrients. Recent studies demonstrate the importance of mTORC1 signaling in β cells. This review will discuss recent advances in the understanding of how this pathway regulates β-cell mass and present data on the role of TSC1 in modulation of β-cell mass. Herein, we also demonstrate that deletion of Tsc1 in pancreatic β cells results in improved glucose tolerance, hyperinsulinemia and expansion of β-cell mass that persists with aging.


Journal of Clinical Investigation | 2014

Maternal diet–induced microRNAs and mTOR underlie β cell dysfunction in offspring

Emilyn U. Alejandro; Brigid Gregg; Taylor Wallen; Doga Kumusoglu; Daniel L. Meister; Angela Chen; Matthew J. Merrins; Leslie S. Satin; Ming Liu; Peter Arvan; Ernesto Bernal-Mizrachi

A maternal diet that is low in protein increases the susceptibility of offspring to type 2 diabetes by inducing long-term alterations in β cell mass and function. Nutrients and growth factor signaling converge through mTOR, suggesting that this pathway participates in β cell programming during fetal development. Here, we revealed that newborns of dams exposed to low-protein diet (LP0.5) throughout pregnancy exhibited decreased insulin levels, a lower β cell fraction, and reduced mTOR signaling. Adult offspring of LP0.5-exposed mothers exhibited glucose intolerance as a result of an insulin secretory defect and not β cell mass reduction. The β cell insulin secretory defect was distal to glucose-dependent Ca2+ influx and resulted from reduced proinsulin biosynthesis and insulin content. Islets from offspring of LP0.5-fed dams exhibited reduced mTOR and increased expression of a subset of microRNAs, and blockade of microRNA-199a-3p and -342 in these islets restored mTOR and insulin secretion to normal. Finally, transient β cell activation of mTORC1 signaling in offspring during the last week of pregnancy of mothers fed a LP0.5 rescued the defect in the neonatal β cell fraction and metabolic abnormalities in the adult. Together, these findings indicate that a maternal low-protein diet alters microRNA and mTOR expression in the offspring, influencing insulin secretion and glucose homeostasis.


Endocrinology | 2010

Acute Insulin Signaling in Pancreatic Beta-Cells Is Mediated by Multiple Raf-1 Dependent Pathways

Emilyn U. Alejandro; Tatyana B. Kalynyak; Farnaz Taghizadeh; Kamila S. Gwiazda; Erin K. Rawstron; Karen J. Jacob; James D. Johnson

Insulin enhances the proliferation and survival of pancreatic beta-cells, but its mechanisms remain unclear. We hypothesized that Raf-1, a kinase upstream of both ERK and Bad, might be a critical target of insulin in beta-cells. To test this hypothesis, we treated human and mouse islets as well as MIN6 beta-cells with multiple insulin concentrations and examined putative downstream targets using immunoblotting, immunoprecipitation, quantitative fluorescent imaging, and cell death assays. Low doses of insulin rapidly activated Raf-1 by dephosphorylating serine 259 and phosphorylating serine 338 in human islets, mouse islets, and MIN6 cells. The phosphorylation of ERK by insulin was eliminated by exposure to a Raf inhibitor (GW5074) or transfection with a dominant-negative Raf-1 mutant. Insulin also enhanced the interaction between mitochondrial Raf-1 and Bcl-2 agonist of cell death (Bad), promoting Bad inactivation via its phosphorylation on serine 112. Insulin-stimulated ERK phosphorylation was abrogated by calcium chelation, calcineurin and calmodulin-dependent protein kinase II inhibitors, and Ned-19, a nicotinic acid adenine dinucleotide phosphate receptor (NAADPR) antagonist. Blocking Raf-1 and Ca(2+) signaling resulted in nonadditive beta-cell death. Autocrine insulin signaling partly accounted for the effects of glucose on ERK phosphorylation. Our results demonstrate that Raf-1 is a critical target of insulin in primary beta-cells. Activation of Raf-1 leads to both an ERK-dependent pathway that involves nicotinic acid adenine dinucleotide phosphate-sensitive Ca(2+) stores and Ca(2+)-dependent phosphorylation events, and an ERK-independent pathway that involves Bad inactivation at the mitochondria. Together our findings identify a novel insulin signaling pathway in beta-cells and shed light on insulins antiapoptotic and mitogenic mechanisms.


Cell Cycle | 2008

Control of pancreatic β-cell fate by insulin signaling: The sweet spot hypothesis

James D. Johnson; Emilyn U. Alejandro

Diabetes results from an absolute or relative deficiency in functional pancreatic β-cell mass. Over the past few years, there has been renewed interest in the role of insulin itself in the regulation of β-cell fate. Numerous animal models point to a critical role for β-cell insulin signaling in the survival and proliferation of pancreatic β-cells. In the present article, we review new studies that elucidate the mechanism by which insulin exerts anti-apoptotic and pro-mitogenic effects on β-cells. In particular, we highlight the emerging role for Raf-1 kinase in autocrine insulin signaling and β-cell fate decisions. We also discuss provocative evidence that the relationship between the dose of insulin and the birth and death of β-cells is not linear. We propose a new hypothesis based on these findings, called the ‘sweet spot’ hypothesis, that can explain how both upward and downward deviations from normal levels of autocrine/paracrine insulin signaling might play an important role in the pathogenesis of type 1 diabetes and type 2 diabetes. We also highlight the key experiments that are required to further test this hypothesis.


The FASEB Journal | 2011

Pancreatic β-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription

Emilyn U. Alejandro; Gareth E. Lim; Arya E. Mehran; Xiaoke Hu; Farnaz Taghizadeh; Dmytro Pelipeychenko; Manuela Baccarini; James D. Johnson

Regulation of glucose homeostasis by insulin depends on pancreatic β‐cell growth, survival, and function. Raf‐1 kinase is a major downstream target of several growth factors that promote proliferation and survival of many cell types, including the pancreatic β cells. We have previously reported that insulin protects β cells from apoptosis and promotes proliferation by activating Raf‐1 signaling in cultured human islets, mouse islets, and MIN6 cells. As Raf‐1 activity is critical for basal apoptosis and insulin secretion in vitro, we hypothesized that Raf‐1 may play an important role in glucose homeostasis in vivo. To test this hypothesis, we utilized the Cre‐loxP recombination system to obtain a pancreatic β‐cell‐specific ablation of Raf‐1 kinase gene (RIPCre+/+: Raf‐1flox/flox) and a complete set of littermate controls (RIPCre+/+:Raf‐1wt/wt). RIPCre+/+:Raf‐1flox/flox mice were viable, and no effects on weight gain were observed. RIPCre+/+:Raf‐1flox/flox mice had increased fasting blood glucose levels and impaired glucose tolerance but normal insulin tolerance compared to littermate controls. Insulin secretion in vivo and in isolated islets was markedly impaired, but there was no apparent effect on the exocytosis machinery. However, islet insulin protein and insulin 2 mRNA, but not insulin 1 mRNA, were dramatically reduced in Raf‐1‐knockout mice. Analysis of insulin 2 knockout mice demonstrated that this reduction in mRNA was sufficient to impair in vivo insulin secretion. Our data further indicate that Raf‐1 specifically and acutely regulates insulin 2 mRNA via negative action on Foxo1, which has been shown to selectively control the insulin 2 gene. This work provides the first direct evidence that Raf‐1 signaling is essential for the regulation of basal insulin transcription and the supply of releasable insulin in vivo.—Alejandro, E. U., Lim, G. E., Mehran, A. E., Hu, X., Taghizadeh, F., Pelipeychenko, D., Baccarini, M., Johnson, J. D. Pancreatic β‐cell Raf‐1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription. FASEB J. 25, 3884–3895 (2011). www.fasebj.org

Collaboration


Dive into the Emilyn U. Alejandro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

James D. Johnson

University of British Columbia

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seokwon Jo

University of Minnesota

View shared research outputs
Researchain Logo
Decentralizing Knowledge