Network


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

Hotspot


Dive into the research topics where Simon T. Hui is active.

Publication


Featured researches published by Simon T. Hui.


Cell Metabolism | 2012

Thioredoxin-interacting protein mediates ER stress-induced β cell death through initiation of the inflammasome.

Christine M. Oslowski; Takashi Hara; Bryan O'Sullivan-Murphy; Kohsuke Kanekura; Simin Lu; Mariko Hara; Shinsuke Ishigaki; Lihua Julie Zhu; Emiko Hayashi; Simon T. Hui; Dale L. Greiner; Randal J. Kaufman; Rita Bortell; Fumihiko Urano

Recent clinical and experimental evidence suggests that endoplasmic reticulum (ER) stress contributes to the life-and-death decisions of β cells during the progression of type 1 and type 2 diabetes. Although crosstalk between inflammation and ER stress has been suggested to play a significant role in β cell dysfunction and death, a key molecule connecting ER stress to inflammation has not been identified. Here we report that thioredoxin-interacting protein (TXNIP) is a critical signaling node that links ER stress and inflammation. TXNIP is induced by ER stress through the PERK and IRE1 pathways, induces IL-1β mRNA transcription, activates IL-1β production by the NLRP3 inflammasome, and mediates ER stress-mediated β cell death. Collectively, our results suggest that TXNIP is a potential therapeutic target for diabetes and ER stress-related human diseases such as Wolfram syndrome.


Cell Metabolism | 2013

Genetic Control of Obesity and Gut Microbiota Composition in Response to High-Fat, High-Sucrose Diet in Mice

Brian W. Parks; Elizabeth Nam; Elin Org; Emrah Kostem; Frode Norheim; Simon T. Hui; Calvin Pan; Mete Civelek; Christoph Rau; Brian J. Bennett; Margarete Mehrabian; Luke K. Ursell; Aiqing He; Lawrence W. Castellani; Bradley A. Zinker; Mark S. Kirby; Thomas A. Drake; Christian A. Drevon; Rob Knight; Peter S. Gargalovic; Todd G. Kirchgessner; Eleazar Eskin; Aldons J. Lusis

Obesity is a highly heritable disease driven by complex interactions between genetic and environmental factors. Human genome-wide association studies (GWAS) have identified a number of loci contributing to obesity; however, a major limitation of these studies is the inability to assess environmental interactions common to obesity. Using a systems genetics approach, we measured obesity traits, global gene expression, and gut microbiota composition in response to a high-fat/high-sucrose (HF/HS) diet of more than 100 inbred strains of mice. Here we show that HF/HS feeding promotes robust, strain-specific changes in obesity that are not accounted for by food intake and provide evidence for a genetically determined set point for obesity. GWAS analysis identified 11 genome-wide significant loci associated with obesity traits, several of which overlap with loci identified in human studies. We also show strong relationships between genotype and gut microbiota plasticity during HF/HS feeding and identify gut microbial phylotypes associated with obesity.


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

Txnip balances metabolic and growth signaling via PTEN disulfide reduction.

Simon T. Hui; Allen M. Andres; Amber K. Miller; Nathanael J. Spann; Douglas W. Potter; Noah M. Post; Amelia Z. Chen; Sowbarnika Sachithanantham; Dae Young Jung; Jason K. Kim; Roger A. Davis

Thioredoxin-interacting protein (Txnip) inhibits thioredoxin NADPH-dependent reduction of protein disulfides. Total Txnip knockout (TKO) mice adapted inappropriately to prolonged fasting by shifting fuel dependence of skeletal muscle and heart from fat and ketone bodies to glucose. TKO mice exhibited increased Akt signaling, insulin sensitivity, and glycolysis in oxidative tissues (skeletal muscle and hearts) but not in lipogenic tissues (liver and adipose tissue). The selective activation of Akt in skeletal muscle and hearts was associated with impaired mitochondrial fuel oxidation and the accumulation of oxidized (inactive) PTEN, whose activity depends on reduction of two critical cysteine residues. Whereas muscle- and heart-specific Txnip knockout mice recapitulated the metabolic phenotype exhibited by TKO mice, liver-specific Txnip knockout mice were similar to WT mice. Embryonic fibroblasts derived from knockout mice also accumulated oxidized (inactive) PTEN and had elevated Akt phosphorylation. In addition, they had faster growth rates and increased dependence on anaerobic glycolysis due to impaired mitochondrial fuel oxidation, and they were resistant to doxorubicin-facilitated respiration-dependent apoptosis. In the absence of Txnip, oxidative inactivation of PTEN and subsequent activation of Akt attenuated mitochondrial respiration, resulting in the accumulation of NADH, a competitive inhibitor of thioredoxin NADPH-reductive activation of PTEN. These findings indicate that, in nonlipogenic tissues, Txnip is required to maintain sufficient thioredoxin NADPH activity to reductively reactivate oxidized PTEN and oppose Akt downstream signaling.


The FASEB Journal | 2008

Thioredoxin-interacting protein deficiency induces Akt/Bcl-xL signaling and pancreatic beta-cell mass and protects against diabetes

Junqin Chen; Simon T. Hui; Francesca M. Couto; Imran N. Mungrue; Dawn Belt Davis; Alan D. Attie; Aldons J. Lusis; Roger A. Davis; Anath Shalev

Pancreatic beta‐cell loss through apoptosis represents a key factor in the pathogenesis of diabetes;however, no effective approaches to block this process and preserve endogenous beta‐cell mass are currently available. To study the role of thioredoxin‐interacting protein (TXNIP), a proapoptotic beta‐cell factor we recently identified, we used HcB‐19 (TXNIP nonsense mutation) and beta‐cell‐specific TXNIP knockout (bTKO) mice. Interestingly, HcB‐19 mice demonstrate increased adiposity, but have lower blood glucose levels and increased pancreatic beta‐cell mass (as assessed by morphometry). Moreover, HcB‐19 mice are resistant to streptozotocin‐induced diabetes. When intercrossed with obese, insulin‐resistant, and diabetic mice, double‐mutant BTBRlepob/obtxniphcb/hcb are even more obese, but are protected against diabetes and beta‐cell apoptosis, resulting in a 3‐fold increase in beta‐cell mass. Beta‐cell‐specific TXNIP deletion also enhanced beta‐cell mass (P< 0.005) and protected against diabetes, and terminal deoxynucleotidyl transferase‐mediated nick end labeling (TUNEL) revealed a ~50‐fold reduction in beta‐cell apoptosis in streptozotocin‐treated bTKO mice. We further discovered that TXNIP deficiency induces Akt/Bcl‐xL signaling and inhibits mitochondrial beta‐cell death, suggesting that these mechanisms may mediate the beta‐cell protective effects of TXNIP deficiency. These results suggest that lowering beta‐cell TXNIP expression could serve as a novel strategy for the treatment of type 1 and type 2 diabetes by promoting endogenous beta‐cell survival.—Chen, J., Hui, S. T., Couto, F. M., Mungrue, I. N., Davis, D. B., Attie, A. D., Lusis, A. J., Davis, R. A., Shalev, A. Thioredoxin‐interacting protein deficiency induces Akt/ Bcl‐xL signaling and pancreatic beta‐cell mass and protects against diabetes. FASEB J. 22, 3581–3594 (2008)


Journal of Biological Chemistry | 2006

Coordinate Transcriptional Repression of Liver Fatty Acid-binding Protein and Microsomal Triglyceride Transfer Protein Blocks Hepatic Very Low Density Lipoprotein Secretion without Hepatosteatosis

Nathanael J. Spann; Sohye Kang; Andrew C. Li; Amelia Z. Chen; Elizabeth P. Newberry; Nicholas O. Davidson; Simon T. Hui; Roger A. Davis

Unlike the livers of humans and mice, and most hepatoma cells, which accumulate triglycerides when treated with microsomal triglyceride transfer protein (MTP) inhibitors, L35 rat hepatoma cells do not express MTP and cannot secrete very low density lipoprotein (VLDL), yet they do not accumulate triglyceride. In these studies we show that transcriptional co-repression of the two lipid transfer proteins, liver fatty acid-binding protein (L-FABP) and MTP, which cooperatively shunt fatty acids into de novo synthesized glycerolipids and the transfer of lipids into VLDL, respectively, act together to maintain hepatic lipid homeostasis. FAO rat hepatoma cells express L-FABP and MTP and demonstrate the ability to assemble and secrete VLDL. In contrast, L35 cells, derived as a single cell clone from FAO cells, do not express L-FABP or MTP nor do they assemble and secrete VLDL. We used these hepatoma cells to elucidate how a conserved DR1 promoter element present in the promoters of L-FABP and MTP affects transcription, expression, and VLDL production. In FAO cells, the DR1 elements of both L-FABP and MTP promoters are occupied by peroxisome proliferator-activated receptor α-retinoid X receptor α (RXRα), with which PGC-1β activates transcription. In contrast, in L35 cells the DR1 elements of both L-FABP and MTP promoters are occupied by chicken ovalbumin upstream promoter transcription factor II, and transcription is diminished. The combined findings indicate that peroxisome proliferator-activated receptor α-RXRα and PGC-1β coordinately up-regulate L-FABP and MTP expression, by competing with chicken ovalbumin upstream promoter transcription factor II for the DR1 sites in the proximal promoters of each gene. Additional studies show that ablation of L-FABP prevents hepatic steatosis caused by treating mice with an MTP inhibitor. Our findings show that reducing both L-FABP and MTP is an effective means to reduce VLDL secretion without causing hepatic steatosis.


Cell Metabolism | 2015

Genetic Architecture of Insulin Resistance in the Mouse

Brian W. Parks; Tamer Sallam; Margarete Mehrabian; Nikolas Psychogios; Simon T. Hui; Frode Norheim; Lawrence W. Castellani; Christoph Rau; Calvin Pan; Jennifer Phun; Zhenqi Zhou; Wen-Pin Yang; Isaac M. Neuhaus; Peter S. Gargalovic; Todd G. Kirchgessner; Mark J. Graham; Richard G. Lee; Peter Tontonoz; Robert E. Gerszten; Andrea L. Hevener; Aldons J. Lusis

Insulin resistance (IR) is a complex trait with multiple genetic and environmental components. Confounded by large differences between the sexes, environment, and disease pathology, the genetic basis of IR has been difficult to dissect. Here we examine IR and related traits in a diverse population of more than 100 unique male and female inbred mouse strains after feeding a diet rich in fat and refined carbohydrates. Our results show dramatic variation in IR among strains of mice and widespread differences between sexes that are dependent on genotype. We uncover more than 15 genome-wide significant loci and validate a gene, Agpat5, associated with IR. We also integrate plasma metabolite levels and global gene expression from liver and adipose tissue to identify metabolite quantitative trait loci (mQTL) and expression QTL (eQTL), respectively. Our results provide a resource for analysis of interactions between diet, sex, and genetic background in IR.


eLife | 2015

The genetic architecture of NAFLD among inbred strains of mice

Simon T. Hui; Brian W. Parks; Elin Org; Frode Norheim; Nam Che; Calvin Pan; Lawrence W. Castellani; Sarada Charugundla; Darwin L. Dirks; Nikolaos Psychogios; Isaac M. Neuhaus; Robert E. Gerszten; Todd G. Kirchgessner; Peter S. Gargalovic; Aldons J. Lusis

To identify genetic and environmental factors contributing to the pathogenesis of non-alcoholic fatty liver disease, we examined liver steatosis and related clinical and molecular traits in more than 100 unique inbred mouse strains, which were fed a diet rich in fat and carbohydrates. A >30-fold variation in hepatic TG accumulation was observed among the strains. Genome-wide association studies revealed three loci associated with hepatic TG accumulation. Utilizing transcriptomic data from the liver and adipose tissue, we identified several high-confidence candidate genes for hepatic steatosis, including Gde1, a glycerophosphodiester phosphodiesterase not previously implicated in triglyceride metabolism. We confirmed the role of Gde1 by in vivo hepatic over-expression and shRNA knockdown studies. We hypothesize that Gde1 expression increases TG production by contributing to the production of glycerol-3-phosphate. Our multi-level data, including transcript levels, metabolite levels, and gut microbiota composition, provide a framework for understanding genetic and environmental interactions underlying hepatic steatosis. DOI: http://dx.doi.org/10.7554/eLife.05607.001


Journal of Biological Chemistry | 2014

Targeted Metabolomics Connects Thioredoxin-interacting Protein (TXNIP) to Mitochondrial Fuel Selection and Regulation of Specific Oxidoreductase Enzymes in Skeletal Muscle

Karen L. DeBalsi; Kari E. Wong; Timothy R. Koves; Dorothy H. Slentz; Sarah E. Seiler; April H. Wittmann; Olga Ilkayeva; Robert D. Stevens; Christopher G. R. Perry; Daniel S. Lark; Simon T. Hui; Luke I. Szweda; P. Darrell Neufer; Deborah M. Muoio

Background: Thioredoxin-interacting protein (TXNIP) is a redox sensor that opposes glucose uptake and glycolytic metabolism. Results: TXNIP-deficient skeletal muscles lose capacity for ketone and branched chain amino acid oxidation due to deficits in specific mitochondrial dehydrogenases. Conclusion: TXNIP permits muscle use of alternative respiratory fuels during glucose deprivation. Significance: Dysregulation of TXNIP might contribute to aberrant fuel selection in the context of metabolic disease. Thioredoxin-interacting protein (TXNIP) is an α-arrestin family member involved in redox sensing and metabolic control. Growing evidence links TXNIP to mitochondrial function, but the molecular nature of this relationship has remained poorly defined. Herein, we employed targeted metabolomics and comprehensive bioenergetic analyses to evaluate oxidative metabolism and respiratory kinetics in mouse models of total body (TKO) and skeletal muscle-specific (TXNIPSKM−/−) Txnip deficiency. Compared with littermate controls, both TKO and TXNIPSKM−/− mice had reduced exercise tolerance in association with muscle-specific impairments in substrate oxidation. Oxidative insufficiencies in TXNIP null muscles were not due to perturbations in mitochondrial mass, the electron transport chain, or emission of reactive oxygen species. Instead, metabolic profiling analyses led to the discovery that TXNIP deficiency causes marked deficits in enzymes required for catabolism of branched chain amino acids, ketones, and lactate, along with more modest reductions in enzymes of β-oxidation and the tricarboxylic acid cycle. The decrements in enzyme activity were accompanied by comparable deficits in protein abundance without changes in mRNA expression, implying dysregulation of protein synthesis or stability. Considering that TXNIP expression increases in response to starvation, diabetes, and exercise, these findings point to a novel role for TXNIP in coordinating mitochondrial fuel switching in response to nutrient availability.


Journal of Lipid Research | 2016

The Hybrid Mouse Diversity Panel: a resource for systems genetics analyses of metabolic and cardiovascular traits.

Aldons J. Lusis; Marcus M. Seldin; Hooman Allayee; Brian J. Bennett; Mete Civelek; Richard C. Davis; Eleazar Eskin; Charles R. Farber; Simon T. Hui; Margarete Mehrabian; Frode Norheim; Calvin Pan; Brian M. Parks; Christoph Rau; Desmond J. Smith; Thomas Q. de Aguiar Vallim; Yibin Wang; Jessica Wang

The Hybrid Mouse Diversity Panel (HMDP) is a collection of approximately 100 well-characterized inbred strains of mice that can be used to analyze the genetic and environmental factors underlying complex traits. While not nearly as powerful for mapping genetic loci contributing to the traits as human genome-wide association studies, it has some important advantages. First, environmental factors can be controlled. Second, relevant tissues are accessible for global molecular phenotyping. Finally, because inbred strains are renewable, results from separate studies can be integrated. Thus far, the HMDP has been studied for traits relevant to obesity, diabetes, atherosclerosis, osteoporosis, heart failure, immune regulation, fatty liver disease, and host-gut microbiota interactions. High-throughput technologies have been used to examine the genomes, epigenomes, transcriptomes, proteomes, metabolomes, and microbiomes of the mice under various environmental conditions. All of the published data are available and can be readily used to formulate hypotheses about genes, pathways and interactions.


Endocrinology | 2013

Induction of the Metabolic Regulator Txnip in Fasting-Induced and Natural Torpor

Laura E. Hand; Ben Saer; Simon T. Hui; H.A. Jinnah; Stephan Steinlechner; Andrew Loudon; David A. Bechtold

Torpor is a physiological state characterized by controlled lowering of metabolic rate and core body temperature, allowing substantial energy savings during periods of reduced food availability or harsh environmental conditions. The hypothalamus coordinates energy homeostasis and thermoregulation and plays a key role in directing torpor. We recently showed that mice lacking the orphan G protein-coupled receptor Gpr50 readily enter torpor in response to fasting and have now used these mice to conduct a microarray analysis of hypothalamic gene expression changes related to the torpor state. This revealed a strong induction of thioredoxin-interacting protein (Txnip) in the hypothalamus of torpid mice, which was confirmed by quantitative RT-PCR and Western blot analyses. In situ hybridization identified the ependyma lining the third ventricle as the principal site of torpor-related expression of Txnip. To characterize further the relationship between Txnip and torpor, we profiled Txnip expression in mice during prolonged fasting, cold exposure, and 2-deoxyglucose-induced hypometabolism, as well as in naturally occurring torpor bouts in the Siberian hamster. Strikingly, pronounced up-regulation of Txnip expression was only observed in wild-type mice when driven into torpor and during torpor in the Siberian hamster. Increase of Txnip was not limited to the hypothalamus, with exaggerated expression in white adipose tissue, brown adipose tissue, and liver also demonstrated in torpid mice. Given the recent identification of Txnip as a molecular nutrient sensor important in the regulation of energy metabolism, our data suggest that elevated Txnip expression is critical to regulating energy expenditure and fuel use during the extreme hypometabolic state of torpor.

Collaboration


Dive into the Simon T. Hui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Calvin Pan

University of California

View shared research outputs
Top Co-Authors

Avatar

Frode Norheim

University of California

View shared research outputs
Top Co-Authors

Avatar

Brian W. Parks

University of California

View shared research outputs
Top Co-Authors

Avatar

Christoph Rau

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian J. Bennett

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jessica Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge