Network


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

Hotspot


Dive into the research topics where Mei Heng Mar is active.

Publication


Featured researches published by Mei Heng Mar.


Developmental Brain Research | 1999

Choline availability alters embryonic development of the hippocampus and septum in the rat

Craig D. Albright; Amy Y. Tsai; Claudia B. Friedrich; Mei Heng Mar; Steven H. Zeisel

Choline availability in the diet during pregnancy alters fetal brain biochemistry with resulting behavioral changes that persist throughout the lifetime of the offspring. In the present study, the effects of dietary choline on cell proliferation, migration, and apoptosis in neuronal progenitor cells in the hippocampus and septum were analyzed in fetal brains at different stages of embryonic development. Timed-pregnant rats on day E12 were fed AIN-76 diet with varying levels of dietary choline for 6 days, and, on days E18 or E20, fetal brain sections were collected. We found that choline deficiency (CD) significantly decreased the rate of mitosis in the neuroepithelium adjacent to the hippocampus. An increased number of apoptotic cells were found in the region of the dentate gyrus of CD hippocampus compared to controls (5.5+/-0.7 vs. 1.9+/-0.3 apoptotic cells per section; p<0.01). Using a combination of bromodeoxyuridine (BrdU) labeling and an unbiased computer-assisted image analysis method, we found that modulation of dietary choline availability changed the distribution and migration of precursor cells born on E16 in the fimbria, primordial dentate gyrus, and Ammons horn of the fetal hippocampus. CD also decreased the migration of newly born cells from the neuroepithelium into the lateral septum, thus indicating that the sensitivity of fetal brain to choline availability is not restricted to the hippocampus. We found an increase in the expression of TOAD-64 protein, an early neuronal differentiation marker, in the hippocampus of CD day E18 fetal brains compared to controls. These results show that dietary choline availability alters the timing of the genesis, migration, and commitment to differentiation of progenitor neuronal-type cells in fetal brain hippocampal regions known to be associated with learning and memory processes in adult brain.


The FASEB Journal | 2003

Homocysteine-betaine interactions in a murine model of 5,10-methylenetetrahydrofolate reductase deficiency

Bernd Schwahn; Zhoutao Chen; Maurice D. Laryea; Udo Wendel; Suzanne Lussier-Cacan; Jacques Genest; Mei Heng Mar; Steven H. Zeisel; Carmen Castro; Timothy A. Garrow; Rima Rozen

Hyperhomocysteinemia, a proposed risk factor for cardiovascular disease, is also observed in other common disorders. The most frequent genetic cause of hyperhomocysteinemia is a mutated methylenetetrahydrofolate reductase (MTHFR), predominantly when folate status is impaired. MTHFR synthesizes a major methyl donor for homocysteine remethylation to methionine. We administered the alternate choline‐derived methyl donor, betaine, to wild‐type mice and to littermates with mild or severe hyperhomocysteinemia due to hetero‐ or homozygosity for a disruption of the Mthfr gene. On control diets, plasma homocysteine and liver choline metabolite levels were strongly dependent on the Mthfr genotype. Betaine supplementation decreased homocysteine in all three genotypes, restored liver betaine and phosphocholine pools, and prevented severe steatosis in Mthfr‐deficient mice. Increasing betaine intake did not further decrease homocysteine. In humans with cardiovascular disease, we found a significant negative correlation between plasma betaine and homocysteine concentrations. Our results emphasize the strong interrelationship between homocysteine, folate, and choline metabolism. Hyperhomocysteinemic Mthfr‐compromised mice appear to be much more sensitive to changes of choline/betaine intake than do wild‐type animals. Hyperhomocysteinemia, in the range of that associated with folate deficiency or with homozygosity for the 677T MTHFR variant, may be associated with disturbed choline metabolism.


The FASEB Journal | 1999

Choline deficiency-induced apoptosis in PC12 cells is associated with diminished membrane phosphatidylcholine and sphingomyelin, accumulation of ceramide and diacylglycerol, and activation of a caspase

Chi Liang E Yen; Mei Heng Mar; Steven H. Zeisel

It is not well appreciated that nutritional status can modulate apoptosis, a process that eliminates unwanted or damaged cells. Choline is an essential nutrient, and its absence induces apoptosis. When PC12 cells were cultivated in a choline‐free medium, apoptosis was induced (27.4% of cells apoptotic at 72 h as compared to 4.4% in control medium). In choline‐free medium at 72 h, there was a 49% decrease in phosphatidylcholine concentration (P<0.01) and a 34% decrease in sphingomyelin concentration (P<0.01); however, there was no change in phosphatidylethanolamine concentration. Before detecting increased apoptosis in choline‐deficient cells, we measured a significant increase in ceramide (218% control) and diacyglycerol (155% control) concentrations. The addition of a cell‐permeable ceramide to cells in control medium induced apoptosis; however, adding a cell‐permeable diacyglycerol did not induce apoptosis. Caspase is a common mediator of apoptosis, and choline deficiency‐induced apoptosis was prevented completely by replacing choline or adding a caspase inhibitor into the medium within 48 h of initial choline deprivation. In those cells rescued by replacing choline at 36 h, the concentrations of phosphatidylcholine, sphingomyelin, ceramide, and diacyglycerol returned to levels of control cells. In those cells rescued by adding a caspase inhibitor at 36 h, the concentrations of sphingomyelin and ceramide returned to control levels, but the concentrations of phosphatidylcholine and diacyglycerol did not return to normal. We propose that availability of dietary factors (choline in this model) can modulate apoptosis. Mechanisms that we identify using this model may help us to explain why dietary choline influences brain development.—Yen, C.‐L. E., Mar, M.‐H., Zeisel, S. H. Choline deficiency‐induced apoptosis in PC12 cells is associated with diminished membrane phosphatidylcholine and sphingomyelin, accumulation of ceramide and diacylglycerol, and activation of a caspase. FASEB J. 13, 135–142 (1999)


Developmental Brain Research | 1999

Maternal dietary choline availability alters mitosis, apoptosis and the localization of TOAD-64 protein in the developing fetal rat septum

Craig D. Albright; Claudia B. Friedrich; Elliott C. Brown; Mei Heng Mar; Steven H. Zeisel

Maternal changes in dietary choline availability alter brain biochemistry and hippocampal development in the offspring resulting in lifelong behavioral changes in the offspring. In order to better understand the relationship between maternal diet, brain cytoarchitecture and behavior, we investigated the effects of choline availability on cell proliferation, apoptosis and differentiation in the fetal rat brain septum. Timed-pregnant rats on day E12 were fed AIN-76 diet with varying levels of dietary choline for 6 days. We found that choline deficiency (CD) significantly decreased the rate of mitosis in the progenitor neuroepithelium adjacent to the septum. In addition, we found an increased number of apoptotic cells in the septum of CD animals compared to controls (3.5+/-0.5 vs. 1.7+/-0.5 apoptotic cells per section; p<0.05). However, CD had no effect on apoptosis in the indusium griseum (IG), a region of cortex dorsal to the septum. Using an unbiased image analysis method and a monoclonal antibody we found a decreased expression of the TOAD-64 kDa protein, a marker of commitment to neuronal differentiation during fetal development, in the dorsal lateral septum of CD animals. CD also decreased the expression of TOAD-64 kDa protein in the IG and cortical plate adjacent to the septum. These results show that dietary choline availability during pregnancy alters the timing of mitosis, apoptosis and the early commitment to neuronal differentiation by progenitor cells in regions of the fetal brain septum, as well as hippocampus, two brain regions known to be associated with learning and memory.


Biochemical Journal | 2003

Phosphatidylethanolamine N-methyltransferase (PEMT) knockout mice have hepatic steatosis and abnormal hepatic choline metabolite concentrations despite ingesting a recommended dietary intake of choline

Xiaonan Zhu; Jiannan Song; Mei Heng Mar; Lloyd J. Edwards; Steven H. Zeisel

Choline is an essential nutrient for humans and is derived from the diet as well as from de novo synthesis involving methylation of phosphatidylethanolamine catalysed by the enzyme phosphatidylethanolamine N -methyltransferase (PEMT). This is the only known pathway that produces new choline molecules. We used mice with a disrupted Pemt-2 gene (which encodes PEMT; Pemt (-/-)) that have previously been shown to possess no hepatic PEMT enzyme. Male, female and pregnant Pemt (-/-) and wild-type mice ( n =5-6 per diet group) were fed diets of different choline content (deficient, control, and supplemented). Livers were collected and analysed for choline metabolites, steatosis, and apoptotic [terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labelling (TUNEL)] positive cells. We found that, in livers of Pemt (-/-) mice fed any of the diets, there was hepatic steatosis and significantly higher occurrence of TUNEL positive cells compared with wild-type controls. In male, female and pregnant mice, liver phosphatidylcholine concentrations were significantly decreased in Pemt (-/-) choline deficient and in Pemt (-/-) choline control groups but returned to normal in Pemt (-/-) choline supplemented groups. Phosphocholine concentrations in liver were significantly diminished in knockout mice even when choline was supplemented to above dietary requirements. These results show that PEMT normally supplies a significant portion of the daily choline requirement in the mouse and, when this pathway is knocked out, mice are unable to attain normal concentrations of all choline metabolites even with a supplemental source of dietary choline.


The FASEB Journal | 2002

Perturbations in choline metabolism cause neural tube defects in mouse embryos in vitro

Melanie C. Fisher; Steven H. Zeisel; Mei Heng Mar; T. W. Sadler

A role for choline during early stages of mammalian embryogenesis has not been established, although recent studies show that inhibitors of choline uptake and metabolism, 2‐dimethylaminoethanol (DMAE), and 1‐O‐octadecyl‐2‐O‐methyl‐rac‐glycero‐3‐phosphocholine (ET‐18‐OCH3), produce neural tube defects in mouse embryos grown in vitro. To determine potential mechanisms responsible for these abnormalities, choline metabolism in the presence or absence of these inhibitors was evaluated in cultured, neurulating mouse embryos by using chromatographic techniques. Results showed that 90%–95% of 14C‐choline was incorporated into phosphocholine and phosphatidylcholine (PtdCho), which was metabolized to sphingomyelin. Choline was oxidized to betaine, and betaine homocysteine methyltransferase was expressed. Acetylcholine was synthesized in yolk sacs, but 70 kDa choline acetyltransferase was undetectable by immunoblot. DMAE reduced embryonic choline uptake and inhibited phosphocholine, PtdCho, phosphatidylethanolamine (PtdEtn), and sphingomyelin synthesis. ET‐18‐OCH3 also inhibited PtdCho synthesis. In embryos and yolk sacs incubated with 3Hethanolamine, 95% of recovered label was PtdEtn, but PtdEtn was not converted to PtdCho, which suggested that phosphatidylethanolamine methyltransferase (PeMT) activity was absent. In ET‐18‐OCH3 treated yolk sacs, PtdEtn was increased, but PtdCho was still not generated through PeMT. Results suggest that endogenous PtdCho synthesis is important during neurulation and that perturbed choline metabolism contributes to neural tube defects produced by DMAE and ET‐18‐OCH3.


The FASEB Journal | 2001

Choline deficiency induces apoptosis in primary cultures of fetal neurons

Chi Liang E Yen; Mei Heng Mar; Rick B. Meeker; Alda Fernandes; Steven H. Zeisel

Treatment of rats with choline during brain development results in long‐lasting enhancement of spatial memory whereas choline deficiency has the opposite effect. Changes in rates of apoptosis may be responsible. We previously demonstrated that choline deficiency induced apoptosis in PC12 cells and suggested that interruption of cell cycling due to a decrease in membrane phosphatidylcholine concentration was the critical mechanism. We now examine whether choline deprivation induces apoptosis in nondividing primary neuronal cultures of fetal rat cortex and hip‐pocampus. Choline deficiency induced widespread apoptosis in primary neuronal cells, indicating that cells do not have to be dividing to be sensitive to choline deficiency. When switched to a choline‐deficient medium, both types of cells became depleted of choline, phosphocholine and phosphatidylcholine, and in primary neurons neurite outgrowth was dramaticallyatten‐uated. Primary cells could be rescued from apoptosis by treatment with phosphocholine or lysophosphatidyl‐choline. As described previously for PC12 cells, an increase in ceramide (Cer) was associated with choline deficiency‐induced apoptosis in primary neurons. The primary neuronal culture appears to be an excellent model to explore the mechanism whereby maternal dietary choline intake modulates apoptosis in the fetal brain.—Yen, C.‐L. E., Mar, M.‐H., Meeker, R. B., Fernandes, A., Zeisel, S. H. Choline deficiency induces apoptosis in primary cultures of fetal neurons. FASEB J. 15, 1704–1710 (2001)


Journal of Cellular Biochemistry | 2003

Mitochondrial and microsomal derived reactive oxygen species mediate apoptosis induced by transforming growth factor-β1 in immortalized rat hepatocytes†

Craig D. Albright; Rudolf I. Salganik; Corneliu N. Craciunescu; Mei Heng Mar; Steven H. Zeisel

Transforming growth factor‐β1 (TGFβ1) is a multifunctional cytokine that is over expressed during liver hepatocytes injury and regeneration. SV40‐transformed CWSV‐1 rat hepatocytes that are p53‐defective undergo apoptosis in response to choline deficiency (CD) or TGFβ1, which mediates CD‐apoptosis. Reactive oxygen species (ROS) are essential mediators of apoptosis. We have shown that apoptosis induced by TGFβ1 is accompanied by ROS generation and the ROS‐trapping agent N‐acetylcysteine (NAC) inhibits TGFβ1‐induced apoptosis. While persistent induction of ROS contributes to this form of apoptosis, the source of ROS generated downstream of TGFβ1 is not clear. The mitochondria and the endoplasmic reticulum both harbor potent electron transfer chains that might be the source of ROS essential for completion of TGFβ1‐apoptosis. Here we show that CWSV‐1 cells treated with cyclosporine A, which prevents opening of mitochondrial membrane pores required for ROS generation, inhibits TGFβ1‐induced apoptosis. A similar effect was obtained by treating these cells with rotenone, an inhibitor of complex 1 of the mitochondrial electron transfer chain. However, we demonstrate that TGFβ1 induces cytochrome P450 1A1 and that metyrapone, a potent inhibitor of cytochrome P450 1A1, inhibits TGFβ1‐induced apoptosis. Therefore, our studies indicate that concurrent with promoting generation of ROS from mitochondria, TGFβ1 also promotes generation of ROS from the cytochrome P450 electron transfer chain. Since inhibition of either of these two sources of ROS interferes with apoptosis, it is reasonable to conclude that the combined involvement of both pathways is essential for completion of TGFβ1‐induced apoptosis. J. Cell. Biochem. 89: 254–261, 2003.


Journal of Cellular Biochemistry | 1997

Methyl-group donors cannot prevent apoptotic death of rat hepatocytes induced by choline-deficiency

Ok Ho Shin; Mei Heng Mar; Craig D. Albright; Maria T. Citarella; Kerry Ann Da Costa; Steven H. Zeisel

Choline‐deficiency causes liver cells to die by apoptosis, and it has not been clear whether the effects of choline‐deficiency are mediated by methyl‐deficiency or by lack of choline moieties. SV40 immortalized CWSV‐1 hepatocytes were cultivated in media that were choline‐sufficient, choline‐deficient, choline‐deficient with methyl‐donors (betaine or methionine), or choline‐deficient with extra folate/vitamin B12. Choline‐deficient CWSV‐1 hepatocytes were not methyl‐deficient as they had increased intracellular S‐adenosylmethionine concentrations (132% of control; P < 0.01). Despite increased phosphatidylcholine synthesis via sequential methylation of phosphatidylethanolamine, choline‐deficient hepatocytes had significantly decreased (P < 0.01) intracellular concentrations of choline (20% of control), phosphocholine (6% of control), glycerophosphocholine (15% of control), and phosphatidylcholine (55% of control). Methyl‐supplementation in choline‐deficiency enhanced intracellular methyl‐group availability, but did not correct choline‐deficiency induced abnormalities in either choline metabolite or phospholipid content in hepatocytes. Methyl‐supplemented, choline‐deficient cells died by apoptosis. In a rat study, 2 weeks of a choline‐deficient diet supplemented with betaine did not prevent the occurrence of fatty liver and the increased DNA strand breakage induced by choline‐deficiency. Though dietary supplementation with betaine restored hepatic betaine concentration and increased hepatic S‐adenosylmethionine/S‐adenosylhomocysteine ratio, it did not correct depleted choline (15% of control), phosphocholine (6% control), or phosphatidylcholine (48% of control) concentrations in deficient livers. These data show that decreased intracellular choline and/or choline metabolite concentrations, and not methyl deficiency, are associated with apoptotic death of hepatocytes. J. Cell. Biochem, 64:196–208.


Cellular Physiology and Biochemistry | 2005

Regulation of choline deficiency apoptosis by epidermal growth factor in CWSV-1 rat hepatocytes.

Craig D. Albright; Kerry Ann Da Costa; Corneliu N. Craciunescu; Erich Klem; Mei Heng Mar; Steven H. Zeisel

Previous studies show that acute choline deficiency (CD) triggers apoptosis in cultured rat hepatocytes (CWSV-1 cells). We demonstrate that prolonged EGF stimulation (10 ng/mL x 48 hrs) restores cell proliferation, as assessed by BrdU labeling, and protects cells from CD-induced apoptosis, as assessed by TUNEL labeling and cleavage of poly(ADP-ribose) polymerase. However, EGF rescue was not accompanied by restoration of depleted intracellular concentrations of choline, glycerphosphocholine, phosphocholine, or phosphatidylcholine. In contrast, we show that EGF stimulation blocks apoptosis by restoring mitochondrial membrane potential (Δ Ψm), as determined using the potential-sensitive dye chloromethyl-X-rosamine, and by preventing the release and nuclear localization of cytochrome c. We investigated whether EGF rescue involves EGF receptor phosphorylation and activation of the down-stream cell survival factor Akt. Compared to cells in control medium (CT, 70 μmol choline x 48hrs), cells in CD medium (5 μmol choline) were less sensitive to EGF-induced (0-300 ng/mL x 5 min) receptor tyrosine phosphorylation. Compared to cells in CT medium, cells in CD medium treated with EGF (10 ng/mL x 5 min) exhibited higher levels of phosphatidylinositol 3-kinase (PI3K)-dependent phosphorylation of AktSer473. Inactivation of PI3K was sufficient to block EGF-stimulated activation of Akt, restoration of mitochondrial Δ Ψm, and prevention of cytochrome c release. These studies indicate that stimulation with EGF activates a cell survival response against CD-apoptosis by restoring mitochondrial membrane potential and preventing cytochrome c release and nuclear translocation which are mediated by activation of Akt in hepatocytes.

Collaboration


Dive into the Mei Heng Mar's collaboration.

Top Co-Authors

Avatar

Steven H. Zeisel

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Craig D. Albright

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Corneliu N. Craciunescu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chi Liang E Yen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jiannan Song

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Kerry Ann Da Costa

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Claudia B. Friedrich

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Elliott C. Brown

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Amy Y. Tsai

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joanne M. Holden

Agricultural Research Service

View shared research outputs
Researchain Logo
Decentralizing Knowledge