Network


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

Hotspot


Dive into the research topics where Maggie M.-Y. Chi is active.

Publication


Featured researches published by Maggie M.-Y. Chi.


Nature Medicine | 1998

Hyperglycemia induces apoptosis in pre-implantation embryos through cell death effector pathways

Kelle H. Moley; Maggie M.-Y. Chi; C.M. Knudson; Stanley J. Korsmeyer; Mike Mueckler

Although perinatal mortality rates have improved for pregnant diabetic women because of insulin therapy and tight metabolic control, infants of diabetics still experience significantly higher rates of congenital malformations and spontaneous miscarriages compared with those of non-diabetic women. Our results here indicate that hyperglycemic conditions, either in vivo or in vitro, modulate the expression of an apoptosis regulatory gene as early as the pre-implantation blastocyst stage in the mouse. Apoptosis in the mammalian pre-implantation blastocyst is a normal process, thought to protect the early embryo by eliminating abnormal cells2. Here we demonstrate that expression of Bax, a Bcl-2-like protein, is increased at the blastocyst stage in the presence of high concentrations of glucose, and that these changes correlate morphologically with increased DNA fragmentation. Expression of Bax and caspase are necessary for this in vitro glucose-induced apoptotic event, and ceramide is involved in mediating this embryotoxic effect of glucose. We also show that these apoptotic cellular changes can be prevented in vivo by treating hyperglycemic mice with insulin before and immediately after conception. These findings emphasize the importance of tight glycemic control in diabetic women at the earliest stages after conception.


PLOS ONE | 2012

High Fat Diet Induced Developmental Defects in the Mouse: Oocyte Meiotic Aneuploidy and Fetal Growth Retardation/Brain Defects

Kerri M. Luzzo; Qiang Wang; Scott H. Purcell; Maggie M.-Y. Chi; Patricia T. Jimenez; Natalia M. Grindler; Tim Schedl; Kelle H. Moley

Background Maternal obesity is associated with poor outcomes across the reproductive spectrum including infertility, increased time to pregnancy, early pregnancy loss, fetal loss, congenital abnormalities and neonatal conditions. Furthermore, the proportion of reproductive-aged woman that are obese in the population is increasing sharply. From current studies it is not clear if the origin of the reproductive complications is attributable to problems that arise in the oocyte or the uterine environment. Methodology/Principal Findings We examined the developmental basis of the reproductive phenotypes in obese animals by employing a high fat diet mouse model of obesity. We analyzed very early embryonic and fetal phenotypes, which can be parsed into three abnormal developmental processes that occur in obese mothers. The first is oocyte meiotic aneuploidy that then leads to early embryonic loss. The second is an abnormal process distinct from meiotic aneuploidy that also leads to early embryonic loss. The third is fetal growth retardation and brain developmental abnormalities, which based on embryo transfer experiments are not due to the obese uterine environment but instead must be from a defect that arises prior to the blastocyst stage. Conclusions/Significance Our results suggest that reproductive complications in obese females are, at least in part, from oocyte maternal effects. This conclusion is consistent with IVF studies where the increased pregnancy failure rate in obese women returns to the normal rate if donor oocytes are used instead of autologous oocytes. We postulate that preconceptional weight gain adversely affects pregnancy outcomes and fetal development. In light of our findings, preconceptional counseling may be indicated as the preferable, earlier target for intervention in obese women desiring pregnancy and healthy outcomes.


Journal of Biological Chemistry | 1998

Glucosamine-induced insulin resistance in 3T3-L1 adipocytes is caused by depletion of intracellular ATP.

Richard C. Hresko; Harry Heimberg; Maggie M.-Y. Chi; Mike Mueckler

Glucosamine, which enters the hexosamine pathway downstream of the rate-limiting step, has been routinely used to mimic the insulin resistance caused by high glucose and insulin. We investigated the effect of glucosamine on insulin-stimulated glucose transport in 3T3-L1 adipocytes. The Δ-insulin (insulin-stimulated minus basal) value for 2-deoxyglucose uptake was dramatically inhibited with increasing concentrations of glucosamine with an ED50 of 1.95 mm. Subcellular fractionation experiments demonstrated that reduction in insulin-stimulated 2-deoxyglucose uptake by glucosamine was due to an inhibition of translocation of both Glut 1 and Glut 4 from the low density microsomes (LDM) to the plasma membrane. Analysis of the insulin signaling cascade revealed that glucosamine impaired insulin receptor autophosphorylation, insulin receptor substrate (IRS-1) phosphorylation, IRS-1-associated PI 3-kinase activity in the LDM, and AKT-1 activation by insulin. Measurement of intracellular ATP demonstrated that the effects of glucosamine were highly correlated with its ability to reduce ATP levels. Reduction of intracellular ATP using azide inhibited Glut 1 and Glut 4 translocation from the LDM to the plasma membrane, insulin receptor autophosphorylation, and IRS-1 tyrosine phosphorylation. Additionally, both the reduction in intracellular ATP and the effects on insulin action caused by glucosamine could be prevented by the addition of inosine, which served as an alternative energy source in the medium. We conclude that direct administration of glucosamine can rapidly lower cellular ATP levels and affect insulin action in fat cells by mechanisms independent of increased intracellular UDP-N-acetylhexosamines and that increased metabolism of glucose via the hexosamine pathway may not represent the mechanism of glucose toxicity in fat cells.


Endocrinology | 2000

High insulin-like growth factor 1 (IGF-1) and insulin concentrations trigger apoptosis in the mouse blastocyst via down-regulation of the IGF-1 receptor.

Maggie M.-Y. Chi; Amanda Schlein; Kelle H. Moley

Women with polycystic ovary syndrome have significantly higher rates of pregnancy loss, as well as elevated insulin and IGF-1 levels. In this study, preimplantation embryos exposed to high concentrations of IGF-1 or insulin undergo extensive apoptosis of the ICM nuclei. Lack of BAX expression, the caspase inhibitor, zVAD, or the ceramide synthase inhibitor, fumonisin B1, prevents this event, suggesting involvement of programmed cell death effector pathways. In other systems, the IGF-1 concentration regulates IGF-1R expression and thus high concentrations lead to down-regulation of the receptor. Here, data show a decrease in IGF-1 receptor protein expression, both by confocal immunofluorescent microscopy and by Western analysis upon exposure to 130 nM IGF-1. Insulin-stimulated glucose uptake, an event regulated via the IGF-1 receptor, is decreased upon exposure to excess IGF-1, suggesting decreased function of the receptor. The data also show that, by blocking receptor signal transduction or by decreasing receptor expression, the apoptotic event can be recreated, thus strongly suggesting that the mechanism of high IGF-1 induced apoptosis is decreased downstream IGF-1 receptor signaling. This embryotoxic insult by high IGF-1 levels may be responsible for the high incidence of pregnancy loss seen in women with polycystic ovary syndrome.


American Journal of Physiology-endocrinology and Metabolism | 1998

Maternal hyperglycemia alters glucose transport and utilization in mouse preimplantation embryos

Kelle H. Moley; Maggie M.-Y. Chi; Mike Mueckler

Glucose utilization was studied in preimplantation embryos from normal and diabetic mice. With use of ultramicrofluorometric enzyme assays, intraembryonic free glucose in single embryos recovered from control and streptozotocin-induced hyperglycemic mice was measured at 24, 48, 72, and 96 h after mating. Free glucose concentrations dropped significantly in diabetics at 48 and 96 h, corresponding to the two-cell and blastocyst stages (48 h: diabetic 0.23 +/- 0.09 vs. control 2.30 +/- 0.43 mmol/kg wet wt; P < 0.001; 96 h: diabetic 0.31 +/- 0.29 vs. control 5.12 +/- 0.17 mmol/kg wet wt; P < 0.001). Hexokinase activity was not significantly different in the same groups. Transport was then compared using nonradioactive 2-deoxyglucose uptake and microfluorometric enzyme assays. The 2-deoxyglucose uptake was significantly lower at both 48 and 96 h in embryos from diabetic vs. control mice (48 h diabetic, 0.037 +/- 0. 003; control, 0.091 +/- 0.021 mmol . kg wet wt-1 . 10 min-1, P < 0. 05; 96 h diabetic, 0.249 +/- 0.008; control, 0.389 +/- 0.007 mmol . kg wet wt-1 . 10 min-1, P < 0.02). When competitive quantitative reverse transcription-polymerase chain reaction was used, there was 44 and 68% reduction in the GLUT-1 mRNA at 48 h (P < 0.001) and 96 h (P < 0.05), respectively, in diabetic vs. control mice. GLUT-2 and GLUT-3 mRNA values were decreased 63 and 77%, respectively (P < 0.01, P < 0.01) at 96 h. Quantitative immunofluorescence microscopy demonstrated 49 +/- 6 and 66 +/- 4% less GLUT-1 protein at 48 and 96 h and 90 +/- 5 and 84 +/- 6% less GLUT-2 and -3 protein, respectively, at 96 h in diabetic embryos. These findings suggest that, in response to a maternal diabetic state, preimplantation mouse embryos experience a decrease in glucose utilization directly related to a decrease in glucose transport at both the mRNA and protein levels.


Journal of Neurochemistry | 1983

Diversity of Metabolic Patterns in Human Brain Tumors: Enzymes of Energy Metabolism and Related Metabolites and Cofactors

Oliver H. Lowry; Sosamma J. Berger; Joyce G. Carter; Maggie M.-Y. Chi; Jill K. Manchester; Joseph Knor; Mary Ellen Pusateri

Abstract: Biopsies from 15 human gliomas, five meningiomas, four Schwannomas, one medulloblastoma, and four normal brain areas were analyzed for 12 enzymes of energy metabolism and 12 related metabolites and cofactors. Samples, 0.01–0.25 μg dry weight, were dissected from freeze‐dried microtome sections to permit all the assays on a given specimen to be made, as far as possible, on nonnecrotic pure tumor tissue from the same region. Great diversity was found with regard to both enzyme activities and metabolite levels among individual tumors, but the following generalities can be made. Activities of hexokinase, phosphorylase, phosphofructokinase, glycerophosphate dehydrogenase, citrate synthase, and malate dehydrogenase levels were usually lower than in brain; glycogen synthase and glucose‐6‐phosphate dehydrogenase were usually higher; and the averages for pyruvate kinase, lactate dehydrogenase, 6‐phosphogluconate dehydrogenase, and β‐hydroxyacyl coenzyme A dehydrogenase were not greatly different from brain. Levels of eight of the 12 enzymes were distinctly lower among the Schwannomas than in the other two groups. Average levels of glucose‐6‐phosphate, lactate, pyruvate, and uridine diphosphoglucose were more than twice those of brain; 6‐phosphogluconate and citrate were about 70% higher than in brain; glucose, glycogen, glycerol‐1‐phosphate, and malate averages ranged from 104% to 127% of brain; and fructose‐1,6‐bisphosphate and glucose‐1,6‐bis‐phosphate levels were on the average 50% and 70% those of brain, respectively.


Journal of Neurochemistry | 1977

Diversity of metabolic patterns in human brain tumors--I. High energy phosphate compounds and basic composition.

Oliver H. Lowry; Sosamma J. Berger; Maggie M.-Y. Chi; Joyce G. Carter; A. Blackshaw; W. Outlaw

Abstract— A total of 25 human brain tumors and 4 specimens of human brain were rapidly frozen at the time of operation and analyzed for ATP, ADP, AMP, UTP, total nucleoside triphosphates, P‐creatine, creatine, inorganic P, creatine kinase, lipid and glycogen. Analyses were made on submicrogram samples dissected from frozen dried sections in order to obtain material as free as possible from admixture with brain, necrotic tissue, blood, etc. A method was developed to estimate the original water content of the frozen dried samples. The brain specimens contained five times as much glycogen as small mammal brains, otherwise the values were similar. The tumors were in fair to excellent energy status. Within the areas chosen for assay, most of ATP and total adenylate were substantially higher than in brain in the case of 5 out of 15 gliomas, 3 of 5 meningiomas, and 1 of 4 schwannomas. UTP was almost invariably higher and other nucleotide triphosphates (besides ATP and UTP) lower than in brain. Glycogen was extremely variable, ranging among the gliomas from 0.05% to 6% of dry wt (4 times the level in the human brains). Creatine plus P‐creatine, compared to cerebral cortex levels, ranged from 15 to 85% in gliomas, was about 25% in meningiomas and the only medulloblastoma, and varied between 6 and 8% in the schwannomas. P‐Creatine varied more or less in keeping with the energy status. Creatine kinase was exceedingly variable. It was almost zero in the schwannomas, the medulloblastomas, 3 of 5 meningiomas, and 2 of 15 gliomas, whereas in some of the gliomas the activity approached that found in brain.


Journal of Biological Chemistry | 2006

Phosphatidylinositol 3-kinase activity is critical for glucose metabolism and embryo survival in murine blastocysts

Joan K. Riley; Mary O. Carayannopoulos; Amanda Wyman; Maggie M.-Y. Chi; Kelle H. Moley

The phosphatidylinositol 3-kinase (PI3K) signal transduction pathway is a well known mediator of cell growth, proliferation, and survival signals. Whereas the expression and function of this pathway has been documented during mammalian development, evidence demonstrating the physiologic importance of this pathway in murine preimplantation embryos is beginning to emerge. This study demonstrates that inhibition of the PI3K pathway leads to the induction of apoptosis in both murine blastocysts and trophoblast stem cells. The apoptosis induced in both model systems correlates with a decrease in the expression of the glucose transporter GLUT1 at the plasma membrane. In addition, blastocysts cultured in the presence of the PI3K inhibitor LY-294002 display a decrease in both 2-deoxyglucose uptake and hexokinase activity as compared with control blastocysts. To determine the impact of PI3K inhibition on pregnancy outcome, embryo transfer experiments were performed. Blastocysts cultured in the presence of LY-294002 demonstrate a dramatic increase in fetal resorptions as compared with control embryos. Finally, we demonstrate that impairment of glucose metabolism via iodoacetate, a glyceraldehyde-3-phosphate dehydrogenase inhibitor, is sufficient to induce apoptosis in both blastocysts and trophoblast stem cells. Moreover, blastocysts treated with iodoacetate result in poor pregnancy outcome as determined by embryo transfer experiments. Taken together these data demonstrate the critical importance of the PI3K pathway in preimplantation embryo survival and pregnancy outcome and further emphasize the importance of glucose utilization and metabolism in cell survival pathways.


Science Signaling | 2016

Trehalose inhibits solute carrier 2A (SLC2A) proteins to induce autophagy and prevent hepatic steatosis

Brian J. DeBosch; Monique R. Heitmeier; Allyson L. Mayer; Cassandra B. Higgins; Jan R. Crowley; Thomas E. Kraft; Maggie M.-Y. Chi; Elizabeth P. Newberry; Zhouji Chen; Brian N. Finck; Nicholas O. Davidson; Kevin E. Yarasheski; Paul W. Hruz; Kelle H. Moley

The disaccharide trehalose blocks glucose uptake in hepatocytes and induces autophagy that prevents fatty liver disease. A sugary inhibitor of liver disease The accumulation of lipids in hepatocytes that occurs in nonalcoholic fatty liver disease (NAFLD) can result in liver failure or liver cancer. Trehalose is a ubiquitous sugar that is present in the food consumed by animals. DeBosch et al. determined that trehalose blocked glucose uptake into cells by inhibiting glucose transporters in the plasma membrane, which induced a “starvation”-like response that activated autophagy even in the presence of adequate nutrients and glucose. Furthermore, providing trehalose to mice that are a model of NAFLD prevented lipid accumulation in the liver. As noted by Mardones et al. in the associated Focus, trehalose, which has been previously under investigation to treat neurodegenerative diseases characterized by toxic protein aggregates, may be a “silver bullet” for treating diseases resulting from inadequate cellular degradative metabolism. Trehalose is a naturally occurring disaccharide that has gained attention for its ability to induce cellular autophagy and mitigate diseases related to pathological protein aggregation. Despite decades of ubiquitous use as a nutraceutical, preservative, and humectant, its mechanism of action remains elusive. We showed that trehalose inhibited members of the SLC2A (also known as GLUT) family of glucose transporters. Trehalose-mediated inhibition of glucose transport induced AMPK (adenosine 5′-monophosphate–activated protein kinase)–dependent autophagy and regression of hepatic steatosis in vivo and a reduction in the accumulation of lipid droplets in primary murine hepatocyte cultures. Our data indicated that trehalose triggers beneficial cellular autophagy by inhibiting glucose transport.


PLOS ONE | 2010

Mitochondrial Dysfunction and Apoptosis in Cumulus Cells of Type I Diabetic Mice

Qiang Wang; Antonina I. Frolova; Scott H. Purcell; Katie L. Adastra; Erica L. Schoeller; Maggie M.-Y. Chi; Tim Schedl; Kelle H. Moley

Impaired oocyte quality has been demonstrated in diabetic mice; however, the potential pathways by which maternal diabetes exerts its effects on the oocyte are poorly understood. Cumulus cells are in direct contact with the oocyte via gap junctions and provide essential nutrients to support oocyte development. In this study, we investigated the effects of maternal diabetes on the mitochondrial status in cumulus cells. We found an increased frequency of fragmented mitochondria, a decreased transmembrane potential and an aggregated distribution of mitochondria in cumulus cells from diabetic mice. Furthermore, while mitochondrial biogenesis in cumulus cells was induced by maternal diabetes, their metabolic function was disrupted as evidenced by lower ATP and citrate levels. Moreover, we present evidence suggesting that the mitochondrial impairments induced by maternal diabetes, at least in part, lead to cumulus cell apoptosis through the release of cytochrome c. Together the deleterious effects on cumulus cells may disrupt trophic and signaling interactions with the oocyte, contributing to oocyte incompetence and thus poor pregnancy outcomes in diabetic females.

Collaboration


Dive into the Maggie M.-Y. Chi's collaboration.

Top Co-Authors

Avatar

Kelle H. Moley

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Oliver H. Lowry

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Antonina I. Frolova

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Qiang Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Mary O. Carayannopoulos

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jill K. Manchester

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Joyce G. Carter

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Tim Schedl

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Amanda Wyman

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David B. McDougal

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge