GCLM

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 1542 Experts worldwide ranked by ideXlab platform

Terrance J Kavanagh - One of the best experts on this subject based on the ideXlab platform.

  • glutamate cysteine ligase modifier subunit GCLM null mice have increased ovarian oxidative stress and accelerated age related ovarian failure
    Endocrinology, 2015
    Co-Authors: Jinhwan Lim, Isaac Mohar, Terrance J Kavanagh, Brooke N Nakamura, Ulrike Luderer
    Abstract:

    Glutathione (GSH) is the one of the most abundant intracellular antioxidants. Mice lacking the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH. Our prior work showed that GSH plays antiapoptotic roles in ovarian follicles. We hypothesized that GCLM(-/-) mice have accelerated ovarian aging due to ovarian oxidative stress. We found significantly decreased ovarian GSH concentrations and oxidized GSH/oxidized glutathione redox potential in GCLM(-/-) vs GCLM(+/+) ovaries. Prepubertal GCLM(-/-) and GCLM(+/+) mice had similar numbers of ovarian follicles, and as expected, the total number of ovarian follicles declined with age in both genotypes. However, the rate of decline in follicles was significantly more rapid in GCLM(-/-) mice, and this was driven by accelerated declines in primordial follicles, which constitute the ovarian reserve. We found significantly increased 4-hydroxynonenal immunostaining (oxidative lipid damage marker) and significantly increased nitrotyrosine immunostaining (oxidative protein damage marker) in prepubertal and adult GCLM(-/-) ovaries compared with controls. The percentage of small ovarian follicles with increased granulosa cell proliferation was significantly higher in prepubertal and 2-month-old GCLM(-/-) vs GCLM(+/+) ovaries, indicating accelerated recruitment of primordial follicles into the growing pool. The percentages of growing follicles with apoptotic granulosa cells were increased in young adult ovaries. Our results demonstrate increased ovarian oxidative stress and oxidative damage in young GCLM(-/-) mice, associated with an accelerated decline in ovarian follicles that appears to be mediated by increased recruitment of follicles into the growing pool, followed by apoptosis at later stages of follicular development.

  • functional significance of glutamate cysteine ligase modifier for erythrocyte survival in vitro and in vivo
    Cell Death & Differentiation, 2013
    Co-Authors: M Foller, Terrance J Kavanagh, Isaac S Harris, Andrew J Elia, R John, Florian Lang, Tak W Mak
    Abstract:

    Erythrocytes endure constant exposure to oxidative stress. The major oxidative stress scavenger in erythrocytes is glutathione. The rate-limiting enzyme for glutathione synthesis is glutamate–cysteine ligase, which consists of a catalytic subunit (GCLC) and a modifier subunit (GCLM). Here, we examined erythrocyte survival in GCLM-deficient (GCLM−/−) mice. Erythrocytes from GCLM−/− mice showed greatly reduced intracellular glutathione. Prolonged incubation resulted in complete lysis of GCLM−/− erythrocytes, which could be reversed by exogenous delivery of the antioxidant Trolox. To test the importance of GCLM in vivo, mice were treated with phenylhydrazine (PHZ; 0.07 mg/g b.w.) to induce oxidative stress. GCLM−/− mice showed dramatically increased hemolysis compared with GCLM+/+ controls. In addition, PHZ-treated GCLM−/− mice displayed markedly larger accumulations of injured erythrocytes in the spleen than GCLM+/+ mice within 24 h of treatment. Iron staining indicated precipitations of the erythrocyte-derived pigment hemosiderin in kidney tubules of GCLM−/− mice and none in GCLM+/+ controls. In fact, 24 h after treatment, kidney function began to diminish in GCLM−/− mice as evident from increased serum creatinine and urea. Consequently, while all PHZ-treated GCLM+/+ mice survived, 90% of PHZ-treated GCLM−/− mice died within 5 days of treatment. In vitro, upon incubation in the absence or presence of additional oxidative stress, GCLM−/− erythrocytes exposed significantly more phosphatidylserine, a cell death marker, than GCLM+/+ erythrocytes, an effect at least partially due to increased cytosolic Ca2+ concentration. Under resting conditions, GCLM−/− mice exhibited reticulocytosis, indicating that the enhanced erythrocyte death was offset by accelerated erythrocyte generation. GCLM is thus indispensable for erythrocyte survival, in vitro and in vivo, during oxidative stress.

  • increased sensitivity to testicular toxicity of transplacental benzo a pyrene exposure in male glutamate cysteine ligase modifier subunit knockout GCLM mice
    Toxicological Sciences, 2012
    Co-Authors: Brooke N Nakamura, Lisa A. Mcconnachie, Isaac Mohar, Yvonne D Hoang, Gregory W Lawson, Mabel M Cortes, Laura Ortiz, Reshma Patel, Bogdan A Rau, Terrance J Kavanagh
    Abstract:

    Polycyclic aromatic hydrocarbons (PAHs), like benzo[a]pyrene (BaP), are ubiquitous environmental pollutants formed by the incomplete combustion of organic materials. The tripeptide glutathione (GSH) is a major antioxidant and is important in detoxification of PAH metabolites. Mice null for the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH concentrations. We investigated the effects of GCLM deletion alone on male fertility and spermatogenesis and its effect on the sensitivity of male embryos to the transplacental testicular toxicity of BaP. GCLM-/- males had dramatically decreased testicular and epididymal GCL enzymatic activity and total GSH concentrations compared with GCLM+/+ littermates. Ratios of reduced to oxidized GSH were significantly increased in GCLM-/- testes. GSH reductase enzymatic activity was increased in GCLM-/- epididymides. We observed no changes in fertility, testicular weights, testicular sperm head counts, or testicular histology and subtle changes in cauda epididymal sperm counts, motility, and morphology in GCLM-/- compared with GCLM+/+ males. Prenatal exposure to BaP from gestational day 7 to 16 was dose dependently associated with significantly decreased testicular and epididymal weights, testicular and epididymal sperm counts, and with vacuolated seminiferous tubules at 10 weeks of age. GCLM-/- males exposed prenatally to BaP had greater decreases in testicular weights, testicular sperm head counts, epididymal sperm counts, and epididymal sperm motility than GCLM+/+ littermates. These results show no effects of GCLM deletion alone on male fertility and testicular spermatogenesis and subtle epididymal effects but support increased sensitivity of GCLM-/- males to the transplacental testicular toxicity of BaP.

  • lack of maternal glutamate cysteine ligase modifier subunit GCLM decreases oocyte glutathione concentrations and disrupts preimplantation development in mice
    Endocrinology, 2011
    Co-Authors: Brooke N Nakamura, Lisa A. Mcconnachie, Terrance J Kavanagh, Thomas J Fielder, Yvonne D Hoang, Jinhwan Lim, Ulrike Luderer
    Abstract:

    Glutathione (GSH) is the most abundant intracellular thiol and an important regulator of cellular redox status. Mice that lack the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH synthesis. Nicotinamide nucleotide transhydrogenase, an inner mitochondrial membrane protein, catalyzes the interconversion of reduced nicotinamide adenine dinucleotide and reduced nicotinamide adenine dinucleotide phosphate; reduced nicotinamide adenine dinucleotide phosphate is required for reduction of GSH disulfide. Previous work supports roles for GSH in preimplantation development. We hypothesized that GCLM−/− mice have increased preimplantation embryonic mortality and that this effect is enhanced by absence of a functioning Nnt gene. GCLM−/− females produced significantly fewer pups per litter than GCLM+/+ littermates. Numbers of oocytes ovulated in a natural estrous cycle or upon superovulation did not differ by genotype. Fewer uterine implantation sites were observed in the GCLM−/− females. Prepubertal GCLM−/− and GCLM+/+ females were superovulated, then mated overnight with a GCLM+/+ male. At 0.5 d postcoitum, GCLM−/− females had significantly lower percentages of zygotes with two pronuclei and higher percentages of zygotes with one pronucleus than GCLM+/+ or GCLM+/− females. At 3.5 d postcoitum, a significantly lower percentage of blastocyst stage embryos was recovered from uteri of GCLM−/− females than GCLM+/+ females. Embryonic development to the blastocyst stage, but not the two-cell stage, was significantly decreased after in vitro fertilization of oocytes from GCLM−/− females compared with GCLM+/+ females. The Nnt mutation did not enhance the effects of GCLM genotype on female fertility. These results demonstrate critical roles for maternal GSH in supporting normal preimplantation development.

  • Rapid activation of glutamate cysteine ligase following oxidative stress
    Journal of Biological Chemistry, 2010
    Co-Authors: Cecile M. Krejsa, Collin C White, Gary L. Schieven, Christopher C Franklin, Jeffrey A Ledbetter, Terrance J Kavanagh
    Abstract:

    Glutamate cysteine ligase (GCL) catalyzes the rate-limiting step in the formation of the cellular antioxidant glutathione (GSH). The GCL holoenzyme consists of two separately coded proteins, a catalytic subunit (GCLC) and a modifier subunit (GCLM). Both GCLC and GLCM are controlled transcriptionally by a variety of cellular stimuli, including oxidative stress. This study addresses post-translational control of GCL activity, which increased rapidly in human lymphocytes following oxidative stress. Activation of GCL occurred within minutes of treatment and without any change in GCL protein levels and coincided with an increase in the proportion of GCLC in the holoenzyme form. Likewise, GCLM shifted from the monomeric form to holoenzyme and higher molecular weight species. Normal rat tissues also showed a distribution of monomeric and higher molecular weight forms. Neither GCL activation, nor the formation of holoenzyme, required a covalent intermolecular disulfide bridge between GCLC and GCLM. However, in immunoprecipitation studies, a neutralizing epitope associated with enzymatic activity was protected following cellular oxidative stress. Thus, the N-terminal portion of GCLC may undergo a change that stabilizes the GCL holoenzyme. Our results suggest that a dynamic equilibrium exists between low and high activity forms of GCL and is altered by transient oxidative stress. This provides a mechanism for the rapid post-translational activation of GCL and maintenance of cellular GSH homeostasis.

Ulrike Luderer - One of the best experts on this subject based on the ideXlab platform.

  • glutamate cysteine ligase modifier subunit GCLM null mice have increased ovarian oxidative stress and accelerated age related ovarian failure
    Endocrinology, 2015
    Co-Authors: Jinhwan Lim, Isaac Mohar, Terrance J Kavanagh, Brooke N Nakamura, Ulrike Luderer
    Abstract:

    Glutathione (GSH) is the one of the most abundant intracellular antioxidants. Mice lacking the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH. Our prior work showed that GSH plays antiapoptotic roles in ovarian follicles. We hypothesized that GCLM(-/-) mice have accelerated ovarian aging due to ovarian oxidative stress. We found significantly decreased ovarian GSH concentrations and oxidized GSH/oxidized glutathione redox potential in GCLM(-/-) vs GCLM(+/+) ovaries. Prepubertal GCLM(-/-) and GCLM(+/+) mice had similar numbers of ovarian follicles, and as expected, the total number of ovarian follicles declined with age in both genotypes. However, the rate of decline in follicles was significantly more rapid in GCLM(-/-) mice, and this was driven by accelerated declines in primordial follicles, which constitute the ovarian reserve. We found significantly increased 4-hydroxynonenal immunostaining (oxidative lipid damage marker) and significantly increased nitrotyrosine immunostaining (oxidative protein damage marker) in prepubertal and adult GCLM(-/-) ovaries compared with controls. The percentage of small ovarian follicles with increased granulosa cell proliferation was significantly higher in prepubertal and 2-month-old GCLM(-/-) vs GCLM(+/+) ovaries, indicating accelerated recruitment of primordial follicles into the growing pool. The percentages of growing follicles with apoptotic granulosa cells were increased in young adult ovaries. Our results demonstrate increased ovarian oxidative stress and oxidative damage in young GCLM(-/-) mice, associated with an accelerated decline in ovarian follicles that appears to be mediated by increased recruitment of follicles into the growing pool, followed by apoptosis at later stages of follicular development.

  • lack of maternal glutamate cysteine ligase modifier subunit GCLM decreases oocyte glutathione concentrations and disrupts preimplantation development in mice
    Endocrinology, 2011
    Co-Authors: Brooke N Nakamura, Lisa A. Mcconnachie, Terrance J Kavanagh, Thomas J Fielder, Yvonne D Hoang, Jinhwan Lim, Ulrike Luderer
    Abstract:

    Glutathione (GSH) is the most abundant intracellular thiol and an important regulator of cellular redox status. Mice that lack the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH synthesis. Nicotinamide nucleotide transhydrogenase, an inner mitochondrial membrane protein, catalyzes the interconversion of reduced nicotinamide adenine dinucleotide and reduced nicotinamide adenine dinucleotide phosphate; reduced nicotinamide adenine dinucleotide phosphate is required for reduction of GSH disulfide. Previous work supports roles for GSH in preimplantation development. We hypothesized that GCLM−/− mice have increased preimplantation embryonic mortality and that this effect is enhanced by absence of a functioning Nnt gene. GCLM−/− females produced significantly fewer pups per litter than GCLM+/+ littermates. Numbers of oocytes ovulated in a natural estrous cycle or upon superovulation did not differ by genotype. Fewer uterine implantation sites were observed in the GCLM−/− females. Prepubertal GCLM−/− and GCLM+/+ females were superovulated, then mated overnight with a GCLM+/+ male. At 0.5 d postcoitum, GCLM−/− females had significantly lower percentages of zygotes with two pronuclei and higher percentages of zygotes with one pronucleus than GCLM+/+ or GCLM+/− females. At 3.5 d postcoitum, a significantly lower percentage of blastocyst stage embryos was recovered from uteri of GCLM−/− females than GCLM+/+ females. Embryonic development to the blastocyst stage, but not the two-cell stage, was significantly decreased after in vitro fertilization of oocytes from GCLM−/− females compared with GCLM+/+ females. The Nnt mutation did not enhance the effects of GCLM genotype on female fertility. These results demonstrate critical roles for maternal GSH in supporting normal preimplantation development.

  • overexpression of glutamate cysteine ligase protects human cov434 granulosa tumour cells against oxidative and γ radiation induced cell death
    Mutagenesis, 2009
    Co-Authors: Mabel M Corteswanstreet, Erich Giedzinski, Charles L Limoli, Ulrike Luderer
    Abstract:

    Ionizing radiation is toxic to ovarian follicles and can cause infertility. Generation of reactive oxygen species (ROS) has been implicated in the toxicity of ionizing radiation in several cell types. We have shown that depletion of the antioxidant glutathione (GSH) sensitizes follicles and granulosa cells to toxicant-induced apoptosis and that supplementation of GSH is protective. The rate-limiting reaction in GSH biosynthesis is catalysed by glutamate-cysteine ligase (GCL), which consists of a catalytic subunit (GCLC) and a regulatory subunit (GCLM). We hypothesized that overexpression of Gclc or GCLM to increase GSH synthesis would protect granulosa cells against oxidant- and radiation-induced cell death. The COV434 line of human granulosa tumour cells was stably transfected with vectors designed for the constitutive expression of Gclc, GCLM, both Gclc and GCLM or empty vector. GCL protein and enzymatic activity and total GSH levels were significantly increased in the GCL subunit-transfected cells. GCL-transfected cells were resistant to cell killing by treatment with hydrogen peroxide compared to control cells. Cell viability declined less in all the GCL subunit-transfected cell lines 1-8 h after 0.5 mM hydrogen peroxide treatment than in control cells. We next examined the effects of GCL overexpression on responses to ionizing radiation. ROS were measured using a redox-sensitive fluorogenic dye in cells irradiated with 0, 1 or 5 Gy of gamma-rays. There was a dose-dependent increase in ROS within 30 min in all cell lines, an effect that was significantly attenuated in Gcl-transfected cells. Apoptosis, assessed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labelling and activated caspase-3 immunoblotting, was significantly decreased in irradiated Gclc-transfected cells compared to irradiated control cells. Suppression of GSH synthesis in Gclc-transfected cells reversed resistance to radiation. These findings show that overexpression of GCL in granulosa cells can augment GSH synthesis and ameliorate various sequelae associated with exposure to oxidative stress and irradiation.

  • Overexpression of glutamate-cysteine ligase protects human COV434 granulosa tumour cells against oxidative and gamma-radiation-induced cell death
    2009
    Co-Authors: Mabel M. Cortes-wanstreet, Erich Giedzinski, L. Limoli, Ulrike Luderer
    Abstract:

    Ionizing radiation is toxic to ovarian follicles and can cause infertility. Generation of reactive oxygen species (ROS) has been implicated in the toxicity of ionizing radiation in several cell types. We have shown that depletion of the antioxidant glutathione (GSH) sensitizes follicles and granulosa cells to toxicant-induced apoptosis and that supplementation of GSH is protective. The rate-limiting reaction in GSH biosynthesis is catalysed by glutamate– cysteine ligase (GCL), which consists of a catalytic subunit (GCLC) and a regulatory subunit (GCLM). We hypothe-sized that overexpression of Gclc or GCLM to increase GSH synthesis would protect granulosa cells against oxidant-and radiation-induced cell death. The COV434 line of human granulosa tumour cells was stably transfected with vectors designed for the constitutive expression of Gclc

  • gonadotropin regulation of glutamate cysteine ligase catalytic and modifier subunit expression in rat ovary is subunit and follicle stage specific
    American Journal of Physiology-endocrinology and Metabolism, 2005
    Co-Authors: Miyun Tsaiturton, Ulrike Luderer
    Abstract:

    We have observed that levels of the antioxidant glutathione (GSH) and protein levels of the catalytic and modifier subunits of the rate-limiting enzyme in GSH synthesis, GCLc and GCLM, increase in ...

Henry Jay Forman - One of the best experts on this subject based on the ideXlab platform.

  • silencing bach1 alters aging related changes in the expression of nrf2 regulated genes in primary human bronchial epithelial cells
    Archives of Biochemistry and Biophysics, 2019
    Co-Authors: Hongqiao Zhang, Lulu Zhou, Kelvin J A Davies, Henry Jay Forman
    Abstract:

    Nrf2 is the master transcription factor regulating the basal and inducible expression of antioxidant genes. With aging, the basal Nrf2 activity is increased but oxidant/electrophile-enhanced activation of Nrf2 signaling is diminished, and these changes are accompanied by an increased expression of Bach1, a repressor of Nrf2 signaling. In this limited follow-up study, we explored how Bach1 may be involved in aging-related alteration in Nrf2 signaling in primary human bronchial epithelial (HBE) cells. Silencing Bach1 with siRNA increased the basal mRNA expression of Nrf2 regulated genes including glutamate cysteine ligase catalytic (GCLC) and modifier subunit (GCLM), NAD(P)H oxidoreductase 1(NQO-1) and heme oxygenase 1(HO-1), in HBE cells from both young (aged 21-29 years) and older (aged 61-69 years) donors. On the other hand, Bach1 silencing affected the induction of Nrf2-regulated genes differentially in young and older HBE cells. Bach1 silencing significantly enhanced sulforaphane-induced expression of HO-1 but had no effect on that of GCLC, GCLM, and NQO1 in young HBE cells. In contrast, Bach1 silencing enhanced sulforaphane-induced expression of GCLC, GCLM and HO-1 but had no effect on that of NQO-1 in older HBE cells. In conclusion, these results suggest that increased Bach1 contributes to aging-related loss of electrophile-enhanced Nrf2 signaling.

  • association of GCLM 588c t and gclc 129t c promoter polymorphisms of genes coding the subunits of glutamate cysteine ligase with ischemic heart disease development in kazakhstan population
    Disease Markers, 2017
    Co-Authors: Liliya Skvortsova, Henry Jay Forman, Anastasia Perfelyeva, Elmira Khussainova, Alma Mansharipova, Leyla Djansugurova
    Abstract:

    Background. Glutamate cysteine ligase (GCL) is a rate-limiting enzyme in synthesis of glutathione. Evidence suggests that genetic variations in the promoter region of genes coding a catalytic subunit (GCLC -129T/C) and a modifier subunit (GCLM -588C/T) of GCL have a functional impact on their transcriptional activity and were associated with various disorders. Hence, we hypothesize whether these two polymorphic variants of GCLM and GCLC genes are associated with the risk of ischemic heart disease (IHD) development in the population of Kazakhstan. Methods. We evaluated 360 patients with IHD and 341 control subjects. Allele frequencies of studied promoters’ polymorphisms were detected by PCR-RFLP analysis. Multiple logistic regression analysis was applied to assess the risk for different genotypes obtained. Results. The presence of -588T allele in GCLM and -129T allele in GCLC gene genotypes was associated with an increased risk of IHD (GCLM -588T: , ; GCLC -129T: , ) for general ethnically mixed group. Analysis of each ethnical groups separately showed the higher risk tendency for Kazakhs as for GCLM -588T (; ) and as for GCLC -129T (, ). For Russians, statistically differences for two polymorphisms were not observed. Conclusion. The two promoter polymorphisms of GCLM (-588C/T) and GCLC (-128T/C) are associated with an increased risk of IHD in Kazakhstan population.

  • Association of GCLM -588C/T and GCLC -129T/C Promoter Polymorphisms of Genes Coding the Subunits of Glutamate Cysteine Ligase with Ischemic Heart Disease Development in Kazakhstan Population
    Hindawi Limited, 2017
    Co-Authors: Liliya Skvortsova, Henry Jay Forman, Anastasia Perfelyeva, Elmira Khussainova, Alma Mansharipova, Leyla Djansugurova
    Abstract:

    Background. Glutamate cysteine ligase (GCL) is a rate-limiting enzyme in synthesis of glutathione. Evidence suggests that genetic variations in the promoter region of genes coding a catalytic subunit (GCLC -129T/C) and a modifier subunit (GCLM -588C/T) of GCL have a functional impact on their transcriptional activity and were associated with various disorders. Hence, we hypothesize whether these two polymorphic variants of GCLM and GCLC genes are associated with the risk of ischemic heart disease (IHD) development in the population of Kazakhstan. Methods. We evaluated 360 patients with IHD and 341 control subjects. Allele frequencies of studied promoters’ polymorphisms were detected by PCR-RFLP analysis. Multiple logistic regression analysis was applied to assess the risk for different genotypes obtained. Results. The presence of -588T allele in GCLM and -129T allele in GCLC gene genotypes was associated with an increased risk of IHD (GCLM -588T: OR=3.92, p=0.003; GCLC -129T: OR=3.22, p=0.03) for general ethnically mixed group. Analysis of each ethnical groups separately showed the higher risk tendency for Kazakhs as for GCLM -588T (OR=4.79; p=0.03) and as for GCLC -129T (OR=4.79, p=0.03). For Russians, statistically differences for two polymorphisms were not observed. Conclusion. The two promoter polymorphisms of GCLM (-588C/T) and GCLC (-128T/C) are associated with an increased risk of IHD in Kazakhstan population

  • competition of nuclear factor erythroid 2 factors related transcription factor isoforms nrf1 and nrf2 in antioxidant enzyme induction
    Redox biology, 2013
    Co-Authors: Nikolai L Chepelev, Hongqiao Zhang, Henry Jay Forman, Kelvin J A Davies, Skye Mcbride, Andrew J Seal, Todd E Morgan, Caleb E Finch, William G Willmore
    Abstract:

    Although the Nrf2 (nuclear factor-erythroid 2 p45 subunit-related factor 2) regulated expression of multiple antioxidant and cytoprotective genes through the electrophile responsive element (EpRE) is well established, interaction of Nrf2/EpRE with Nrf1, a closely-related transcription factor, is less well understood. Due to either proteolysis or alternative translation, Nrf1 has been found as proteins of varying size, p120, p95, and p65, which have been described as either activators of EpRE or competitive inhibitors of Nrf2. We investigated the effect of Nrf1 on EpRE-regulated gene expression using the catalytic and modifier subunits of glutamate cysteine ligase (GCLC and GCLM) as models and explored the potential role of Nrf1 in altering their expression in aging and upon chronic exposure to airborne nano-sized particulate matter (nPM). Nrf1 knockout resulted in the increased expression of GCLC and GCLM in human bronchial epithelial (HBE1) cells. Overexpression Nrf2 in combination with either p120 or p65 diminished or failed to further increase the GCLC- and GLCM-EpRE luciferase activity. All known forms of Nrf1 protein, remained unchanged in the lungs of mice with age or in response to nPM. Our study shows that Nrf1 could inhibit EpRE activity in vitro, whereas the precise role of Nrf1 in vivo requires further investigations. We conclude that Nrf1 may not be directly responsible for the loss of Nrf2-dependent inducibility of antioxidant and cytoprotective genes observed in aged animals.

  • Structure, function, and post-translational regulation of the catalytic and modifier subunits of glutamate cysteine ligase.
    Molecular aspects of medicine, 2008
    Co-Authors: Christopher C Franklin, Isaac Mohar, Collin C White, Donald S Backos, Henry Jay Forman, Terrance J Kavanagh
    Abstract:

    Glutathione (GSH) is a tripeptide composed of glutamate, cysteine, and glycine. The first and rate-limiting step in GSH synthesis is catalyzed by glutamate cysteine ligase (GCL, previously known as gamma-glutamylcysteine synthetase). GCL is a heterodimeric protein composed of catalytic (GCLC) and modifier (GCLM) subunits that are expressed from different genes. GCLC catalyzes a unique gamma-carboxyl linkage from glutamate to cysteine and requires ATP and Mg(++) as cofactors in this reaction. GCLM increases the V(max) and K(cat) of GCLC, decreases the K(m) for glutamate and ATP, and increases the K(i) for GSH-mediated feedback inhibition of GCL. While post-translational modifications of GCLC (e.g. phosphorylation, myristoylation, caspase-mediated cleavage) have modest effects on GCL activity, oxidative stress dramatically affects GCL holoenzyme formation and activity. Pyridine nucleotides can also modulate GCL activity in some species. Variability in GCL expression is associated with several disease phenotypes and transgenic mouse and rat models promise to be highly useful for investigating the relationships between GCL activity, GSH synthesis, and disease in humans.

Timothy P Dalton - One of the best experts on this subject based on the ideXlab platform.

  • Early onset senescence occurs when fibroblasts lack the glutamate-cysteine ligase modifier subunit.
    Free Radical Biology and Medicine, 2009
    Co-Authors: Ying Chen, Vasilis Vasiliou, Howard G Shertzer, Elisabet Johansson, Timothy P Dalton
    Abstract:

    Abstract Cellular senescence is the irreversible entry of cells into growth arrest. Senescence of primary cells in culture has long been used as an in vitro model for aging. Glutamate–cysteine ligase (GCL) controls the synthetic rate of the important cellular antioxidant glutathione (GSH). The catalytic subunit of GCL, GCLC, is catalytically active and essential for life. By contrast the modifier subunit of GCL, GCLM, is dispensable in mice. Although it is recognized that GCLM increases the rate of GSH synthesis, its physiological role is unclear. Herein, we show that loss of GCLM leads to premature senescence of primary murine fibroblasts as characterized by: (a) diminished growth rate, (b) cell morphology consistent with senescence, (c) increases in senescence-associated β-galactosidase activity, and (d) cell cycle arrest at the G 1 /S and G 2 /M boundaries. These changes are accompanied by increased intracellular ROS, accumulation of DNA damage, and induction of p53 and p21 proteins. We also found that N- acetylcysteine increases intracellular GSH and prevents premature senescence in GCLM −/− cells. These results suggest that the control of GCLM, which in turn controls aspects of the cellular redox environment via GSH, is important in determining the replicative capacity of the cell.

  • Interaction between the catalytic and modifier subunits of glutamate-cysteine ligase
    Biochemical Pharmacology, 2007
    Co-Authors: Yi Yang, Scott N. Schneider, Howard G Shertzer, Elisabet Johansson, Ying Chen, Timothy P Dalton
    Abstract:

    Abstract Glutamate-cysteine ligase (GCL) is the rate-limiting enzyme in the glutathione (GSH) biosynthesis pathway. This enzyme is a heterodimer, comprising a catalytic subunit (GCLC) and a regulatory subunit (GCLM). Although GCLC alone can catalyze the formation of l -γ-glutamyl- l -cysteine, its binding with GCLM enhances the enzyme activity by lowering the K m for glutamate and ATP, and increasing the K i for GSH inhibition. To characterize the enzyme structure–function relationship, we investigated the heterodimer formation between GCLC and GCLM, in vivo using the yeast two-hybrid system, and in vitro using affinity chromatography. A strong and specific interaction between GCLC and GCLM was observed in both systems. Deletion analysis indicated that most regions, except a portion of the C-terminal region of GCLC and a portion of the N-terminal region of GCLM, are required for the interaction to occur. Point mutations of selected amino acids were also tested for the binding activity. The GCLC Cys248Ala/Cys249Ala and Pro158Leu mutations enzyme showed the same strength of binding to GCLM as did wild-type GCLC, yet the catalytic activity was dramatically decreased. The results suggest that the heterodimer formation may not be dependent on primary amino-acid sequence but, instead, involves a complex formation of the tertiary structure of both proteins.

  • Glutamate Cysteine Ligase Catalysis DEPENDENCE ON ATP AND MODIFIER SUBUNIT FOR REGULATION OF TISSUE GLUTATHIONE LEVELS
    Journal of Biological Chemistry, 2005
    Co-Authors: Ying Chen, Scott N. Schneider, Howard G Shertzer, Timothy P Dalton
    Abstract:

    Abstract Glutamate cysteine ligase (GCL), which synthesizes γ-glutamyl-cysteine (γ-GC), is the rate-limiting enzyme in GSH biosynthesis. γ-GC may be produced by the catalytic subunit GCLC or by the holoenzyme (GCLholo), which comprises GCLC and the modifier subunit GCLM. The GCLM(–/–) knock-out mouse shows tissue levels of GSH that are between 9 and 40% of the GCLM(+/+) wild-type mouse. In the present study, we used recombinant GCLC and GCLM and GCLM(–/–) mice to examine the role of GCLM on γ-GC synthesis by GCLholo. GCLM decreased the Km for ATP by ∼6-fold and, similar to other species, decreased the Km for glutamate and increased the Ki for feedback inhibition by GSH. Furthermore, GCLM increased by 4.4-fold the Kcat for γ-GC synthesis; this difference in catalytic efficiency of GCLholo versus GCLC allowed us to derive a mathematical relationship for γ-GC production and to determine the relative levels of GCLholo and GCLC; in homogenates of brain, liver, and lung, the ratio of GCLC to GCLholo was 7.0, 2.0, and 3.5, respectively. In kidney, however, the relationship between GCLC and GCLholo was complicated. Kidney contains GCLholo, free GCLC, and free GCLM, and free GCLC in kidney cannot interact with GCLM. Taken together, we conclude that, in most tissues, GCLM is limiting, suggesting that an increase in GCLM alone would increase γ-GC synthesis. On the other hand, our results from kidney suggest that γ-GC synthesis may be controlled post-translationally.

  • initial characterization of the glutamate cysteine ligase modifier subunit GCLM knockout mouse novel model system for a severely compromised oxidative stress response
    Journal of Biological Chemistry, 2002
    Co-Authors: Yi Yang, Howard G Shertzer, Ying Chen, Matthew Z Dieter, Daniel W Nebert, Timothy P Dalton
    Abstract:

    Glutamate-cysteine ligase (GCL) is the rate-limiting enzyme in the GSH biosynthesis pathway. In higher eukaryotes, this enzyme is a heterodimer comprising a catalytic subunit (GCLC) and a modifier subunit (GCLM), which change the catalytic characteristics of the holoenzyme. To define the cellular function of GCLM, we disrupted the mouse GCLM gene to create a null allele. GCLM(-/-) mice are viable and fertile and have no overt phenotype. In liver, lung, pancreas, erythrocytes, and plasma, however, GSH levels in GCLM(-/-) mice were 9-16% of that in GCLM(+/+) littermates. Cysteine levels in GCLM(-/-) mice were 9, 35, and 40% of that in GCLM(+/+) mice in kidney, pancreas, and plasma, respectively, but remained unchanged in the liver and erythrocytes. Comparing the hepatic GCL holoenzyme with GCLC in the genetic absence of GCLM, we found the latter had an approximately 2-fold increase in K(m) for glutamate and a dramatically enhanced sensitivity to GSH inhibition. The major decrease in GSH, combined with diminished GCL activity, rendered GCLM(-/-) fetal fibroblasts strikingly more sensitive to chemical oxidants such as H(2)O(2). We conclude that the GCLM(-/-) mouse represents a model of chronic GSH depletion that will be very useful in evaluating the role of the GCLM subunit and GSH in numerous pathophysiological conditions as well as in environmental toxicity associated with oxidant insult.

  • glutamate cysteine ligase modifier subunit mouse GCLM gene structure and regulation by agents that cause oxidative stress
    Biochemical Pharmacology, 2002
    Co-Authors: Willy A Solis, Timothy P Dalton, Howard G Shertzer, Matthew Z Dieter, Sarah L Freshwater, Judy M Harrer, Daniel W Nebert
    Abstract:

    Abstract Glutamate–cysteine ligase is a heterodimer comprising a modifier (GCLM) and a catalytic (GCLC) subunit. In mouse Hepa-1c1c7 hepatoma cell cultures, we found that tert-butylhydroquinone (tBHQ; 50 μM) induces the GCLM and GCLC mRNAs ∼10- and ∼2-fold, respectively, and that these increases primarily reflect de novo transcription. We determined that the mouse GCLM gene has seven exons, spanning 22.3 kb; all exons, intron–exon junctions, and 4.7 kb of 5′-flanking region were sequenced. By RNase protection analysis, we identified two major and several minor transcription start-site clusters over a 300-bp region. The GCLM 5′-flanking region is GC-rich and lacks a canonical TATA box. Transient and stable transfection studies, using luciferase reporter constructs containing incremental GCLM 5′-flanking deletions (4.7–0.5 kb), showed high basal activity but only modest (∼2-fold) inducibility by tBHQ. The only candidate motif for oxidative stress regulation (in the 4.7-kb region we sequenced) is a putative inverted electrophile response element (EPRE) 9 bp upstream from the 5′-most transcription start-site. Site-directed mutagenesis of this −9 EPRE demonstrated minimal (30–40%) decreases in tBHQ induction and no effect on basal activity—suggesting that this EPRE might be necessary but not sufficient. The nuclear erythroid factor-2 (NEF2)-related factor-2 (NRF2) is known to transactivate via EPRE motifs. In the presence of co-transfected NRF cDNA expression vector, however, no increase in GCLM promoter activity was observed. Thus, the endogenous GCLM gene shows robust transcriptional activation by tBHQ in the intact Hepa-1 cell, but reporter constructs containing up to 4.7 kb of promoter (having only the one EPRE at −9) demonstrate a disappointing response, indicating that the major tBHQ-responsive regulatory element of the mouse GCLM gene must exist either further 5′- or 3′-ward of the 4.7-kb region studied.

Christopher C Franklin - One of the best experts on this subject based on the ideXlab platform.

  • Glycation of Glutamate Cysteine Ligase by 2-Deoxy- d -Ribose and its Potential Impact on Chemoresistance in Glioblastoma
    Neurochemical Research, 2013
    Co-Authors: Donald S Backos, Kristofer S. Fritz, Debbie G. Mcarthur, Andrew M. Donson, Nicholas K. Foreman, Jadwiga K Kepa, Christopher C Franklin, Philip Reigan
    Abstract:

    The antioxidant glutathione (GSH) plays a critical role in maintaining intracellular redox homeostasis but in tumors the GSH biosynthetic pathway is often dysregulated, contributing to tumor resistance to radiation and chemotherapy. Glutamate-cysteine ligase (GCL) catalyzes the first and rate-limiting reaction in GSH synthesis, and enzyme function is controlled by GSH feedback inhibition or by transcriptional upregulation of the catalytic (GCLC) and modifier (GCLM) subunits. However, it has recently been reported that the activity of GCLC and the formation of GCL can be modified by reactive aldehyde products derived from lipid peroxidation. Due to the susceptibility of GCLC to posttranslational modifications by reactive aldehydes, we examined the potential for 2-deoxy-d-ribose (2dDR) to glycate GCLC and regulate enzyme activity and GCL formation. 2dDR was found to directly modify both GCLC and GCLM in vitro, resulting in a significant inhibition of GCLC and GCL enzyme activity without altering substrate affinity or feedback inhibition. 2dDR-mediated glycation also inhibited GCL subunit heterodimerization and formation of the GCL holoenzyme complex while not causing dissociation of pre-formed holoenzyme. This PTM could be of particular importance in glioblastoma (GBM) where intratumoral necrosis provides an abundance of thymidine, which can be metabolized by thymidine phosphorylase (TP) to form 2dDR. TP is expressed at high levels in human GBM tumors and shRNA knockdown of TP in U87 GBM cells results in a significant increase in cellular GCL enzymatic activity.

  • Posttranslational modification and regulation of glutamate-cysteine ligase by the α,β-unsaturated aldehyde 4-hydroxy-2-nonenal
    Free Radical Biology and Medicine, 2010
    Co-Authors: Donald S Backos, Kristofer S. Fritz, James R Roede, Christopher C Franklin
    Abstract:

    Abstract 4-Hydroxy-2-nonenal (4-HNE) is a lipid peroxidation product formed during oxidative stress that can alter protein function via adduction of nucleophilic amino acid residues. 4-HNE detoxification occurs mainly via glutathione (GSH) conjugation and transporter-mediated efflux. This results in a net loss of cellular GSH, and restoration of GSH homeostasis requires de novo GSH biosynthesis. The rate-limiting step in GSH biosynthesis is catalyzed by glutamate–cysteine ligase (GCL), a heterodimeric holoenzyme composed of a catalytic (GCLC) and a modulatory (GCLM) subunit. The relative levels of the GCL subunits are a major determinant of cellular GSH biosynthetic capacity and 4-HNE induces the expression of both GCL subunits. In this study, we demonstrate that 4-HNE can alter GCL holoenzyme formation and activity via direct posttranslational modification of the GCL subunits in vitro. 4-HNE directly modified Cys553 of GCLC and Cys35 of GCLM in vitro, which significantly increased monomeric GCLC enzymatic activity, but reduced GCL holoenzyme activity and formation of the GCL holoenzyme complex. In silico molecular modeling studies also indicate these residues are likely to be functionally relevant. Within a cellular context, this novel posttranslational regulation of GCL activity could significantly affect cellular GSH homeostasis and GSH-dependent detoxification during periods of oxidative stress.

  • Rapid activation of glutamate cysteine ligase following oxidative stress
    Journal of Biological Chemistry, 2010
    Co-Authors: Cecile M. Krejsa, Collin C White, Gary L. Schieven, Christopher C Franklin, Jeffrey A Ledbetter, Terrance J Kavanagh
    Abstract:

    Glutamate cysteine ligase (GCL) catalyzes the rate-limiting step in the formation of the cellular antioxidant glutathione (GSH). The GCL holoenzyme consists of two separately coded proteins, a catalytic subunit (GCLC) and a modifier subunit (GCLM). Both GCLC and GLCM are controlled transcriptionally by a variety of cellular stimuli, including oxidative stress. This study addresses post-translational control of GCL activity, which increased rapidly in human lymphocytes following oxidative stress. Activation of GCL occurred within minutes of treatment and without any change in GCL protein levels and coincided with an increase in the proportion of GCLC in the holoenzyme form. Likewise, GCLM shifted from the monomeric form to holoenzyme and higher molecular weight species. Normal rat tissues also showed a distribution of monomeric and higher molecular weight forms. Neither GCL activation, nor the formation of holoenzyme, required a covalent intermolecular disulfide bridge between GCLC and GCLM. However, in immunoprecipitation studies, a neutralizing epitope associated with enzymatic activity was protected following cellular oxidative stress. Thus, the N-terminal portion of GCLC may undergo a change that stabilizes the GCL holoenzyme. Our results suggest that a dynamic equilibrium exists between low and high activity forms of GCL and is altered by transient oxidative stress. This provides a mechanism for the rapid post-translational activation of GCL and maintenance of cellular GSH homeostasis.

  • distinct nrf1 2 independent mechanisms mediate as3 induced glutamate cysteine ligase subunit gene expression in murine hepatocytes
    Free Radical Biology and Medicine, 2009
    Co-Authors: James A Thompson, Terrance J Kavanagh, Nelson Fausto, Charles L White, David P Cox, Jefferson Y Chan, Christopher C Franklin
    Abstract:

    Trivalent arsenite (As(3+)) is a known human carcinogen that is also capable of inducing apoptotic cell death. Increased production of reactive oxygen species is thought to contribute to both the carcinogenic and the cytotoxic effects of As(3+). Glutathione (GSH) constitutes a vital cellular defense mechanism against oxidative stress. The rate-limiting enzyme in GSH biosynthesis is glutamate-cysteine ligase (GCL), a heterodimeric holoenzyme composed of a catalytic (GCLC) and a modifier (GCLM) subunit. In this study, we demonstrate that As(3+) coordinately upregulates Gclc and GCLM mRNA levels in a murine hepatocyte cell line resulting in increased GCL subunit protein expression, holoenzyme formation, and activity. As(3+) increased the rate of transcription of both the GCLM and the Gclc genes and induced the posttranscriptional stabilization of GCLM mRNA. The antioxidant N-acetylcysteine abolished As(3+)-induced Gclc expression and attenuated induction of GCLM. As(3+) induction of Gclc and GCLM was also differentially regulated by the MAPK signaling pathways and occurred independent of the Nrf1/2 transcription factors. These findings demonstrate that distinct transcriptional and posttranscriptional mechanisms mediate the coordinate induction of the Gclc and GCLM subunits of GCL in response to As(3+) and highlight the potential importance of the GSH antioxidant defense system in regulating As(3+)-induced responses in hepatocytes.

  • Structure, function, and post-translational regulation of the catalytic and modifier subunits of glutamate cysteine ligase.
    Molecular aspects of medicine, 2008
    Co-Authors: Christopher C Franklin, Isaac Mohar, Collin C White, Donald S Backos, Henry Jay Forman, Terrance J Kavanagh
    Abstract:

    Glutathione (GSH) is a tripeptide composed of glutamate, cysteine, and glycine. The first and rate-limiting step in GSH synthesis is catalyzed by glutamate cysteine ligase (GCL, previously known as gamma-glutamylcysteine synthetase). GCL is a heterodimeric protein composed of catalytic (GCLC) and modifier (GCLM) subunits that are expressed from different genes. GCLC catalyzes a unique gamma-carboxyl linkage from glutamate to cysteine and requires ATP and Mg(++) as cofactors in this reaction. GCLM increases the V(max) and K(cat) of GCLC, decreases the K(m) for glutamate and ATP, and increases the K(i) for GSH-mediated feedback inhibition of GCL. While post-translational modifications of GCLC (e.g. phosphorylation, myristoylation, caspase-mediated cleavage) have modest effects on GCL activity, oxidative stress dramatically affects GCL holoenzyme formation and activity. Pyridine nucleotides can also modulate GCL activity in some species. Variability in GCL expression is associated with several disease phenotypes and transgenic mouse and rat models promise to be highly useful for investigating the relationships between GCL activity, GSH synthesis, and disease in humans.