51
|
Celik HA, Kircelli F, Saydam G, Aydin HH. Potential involvement of serine/threonine protein phosphatases in apoptosis of HepG2 cells during selenite treatment. Biol Trace Elem Res 2007; 117:65-75. [PMID: 17873393 DOI: 10.1007/bf02698084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 09/05/2006] [Accepted: 10/23/2006] [Indexed: 11/25/2022]
Abstract
Selenium, an essential biological trace element present in both prokaryotic and eukaryotic cells, exerts its regulatory effect in a variety of cellular events, including cell growth, survival, and death. Selenium compounds have been shown in different cell lines to inhibit apoptosis by several mechanisms. Serine/threonine phosphatases (STPs) are potentially important in selenite-induced apoptosis because of their role in regulation of diverse set of cellular processes. In this study, the regulatory role of STPs in selenite-induced apoptosis has been implied by the use of two specific inhibitors: ocadaic acid and calyculin A. Our results show a decrease in cell density in HepG2 cells under selenite treatment. Resulting specific enzyme activities showed a concentration-dependent increase in all three phosphatase activities after 24 h in cells treated with 5 microM selenite and these activities decreased at 48 and 72 h. However, in cells treated with 10 microM selenite, PP2A and PP2B decreased at 48 h, whereas PP2C activity did not change at this dose. In cells treated with 25 microM, there was not a significant change in PP2C activity. These data suggest that the most specific response to selenite treatment was in PP2A and PP2B activities in a dose-dependent manner. Our results with OA and Cal-A further support the view that PP1 and PP2A might act as negative regulators of growth. With these data, we have first demonstrated the role of serine/threonine protein phosphatases in the signaling pathway of selenite-induced apoptosis and resulting cytotoxicity.
Collapse
Affiliation(s)
- Handan Ak Celik
- Department of Biochemistry, School of Medicine, Bornova, Izmir, TR-35100, Turkey
| | | | | | | |
Collapse
|
52
|
Shalini S, Bansal MP. Role of selenium in spermatogenesis: differential expression of cjun and cfos in tubular cells of mice testis. Mol Cell Biochem 2006; 292:27-38. [PMID: 17066317 DOI: 10.1007/s11010-006-9168-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 02/27/2006] [Indexed: 11/26/2022]
Abstract
Selenium (Se) is an essential dietary trace element, involved in the process of male reproduction. Best known as an antioxidant, it acts through various selenoproteins viz. glutathione peroxidase, thioredoxin reductase and selenoprotein P. The aim of the present study was to identify the underlying molecular mechanism of Se in regulating spermatogenesis. Different Se status: deficient, adequate and excess Se, were generated in male Balb/c mice by feeding yeast based Se deficient diet, and deficient diet supplemented with Se as sodium selenite (0.2 and 1 ppm Se) respectively for a period of 4 and 8 weeks. Se levels and glutathione peroxidase (GSH-Px) activity were significantly reduced in the Se deficient mice and enhanced in Se supplemented group. Reduction in the number of post-meiotic germ cells viz. spermatids and spermatozoa, were observed in the deficient groups indicating loss in fertility and reproductive ability. cjun and cfos (components of transcription factor AP1) regulate cellular growth and differentiation and also exert a regulatory role in steroidogenesis and spermatogenesis. Changes in the mRNA expression of cjun and cfos were observed. Concomitant with this, western blot revealed that the protein expression profile for both these genes was significantly altered in the Se deficient and Se excess groups. Further immunohistochemical analysis showed that, both these genes had identical cellular localization indicating that they do not work alone but act synergistically as AP1. cjun and cfos expression was greater in the early mitotic stages-spermatogonia and spermatocytes in the Se adequate controls. It decreased in the meiotic stages and then again peaked around the later stages-elongating spermatids and spermatozoa. However in the Se deficient mice, weaker expression was observed in the spermatogonia with a complete absence of expression near the lumen. No visible changes in cjun/cfos expression and immunohistochemical localization were observed in the excess group compared to the Se adequate controls. In conclusion, the present study clearly demonstrates that alteration in Se supply leads to decreased expression pattern for both cJun and cFos in the testicular germ cells which might be responsible for decreased germ cell number, differentiation and reduced fertility and accounts for the mechanism of Se action in regulating spermatogenesis.
Collapse
Affiliation(s)
- Sonia Shalini
- Department of Biophysics, Panjab University, Chandigarh 160014, India
| | | |
Collapse
|
53
|
Park HS, Huh SH, Kim MS, Kim DY, Gwag BJ, Cho SG, Choi EJ. Neuronal nitric oxide synthase (nNOS) modulates the JNK1 activity through redox mechanism: a cGMP independent pathway. Biochem Biophys Res Commun 2006; 346:408-14. [PMID: 16764826 DOI: 10.1016/j.bbrc.2006.05.122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 05/14/2006] [Indexed: 11/26/2022]
Abstract
Nitric oxide (NO) is a small, uncharged molecule, which is primarily generated by the nitric oxide synthase (NOS) family of proteins, including neuronal nitric oxide synthase (nNOS), inducible NOS (iNOS), and endothelial NOS (eNOS). NO has been implicated in diverse roles in biological systems, such as the regulation of cell death and survival signaling pathways of a variety of cell types, including neuronal cells. In this study, we determined that the NO generated from l-arginine by ectopically overexpressed nNOS in HEK293 cells exerted an inhibitory effect against the activity of c-Jun N-terminal kinase (JNK), an important modulator of neuronal cell death and survival signaling pathways. NO repressed the activation of JNK, but exerted no significant effects on the activities of SEK1/MKK4 and MEKK1, which are the upstream MAPKK and MAPKKK of JNK1, respectively. This NO-mediated inhibition of JNK1 was not affected by the addition of ODQ, a guanylyl cyclase inhibitor, indicating that the effect is independent of the level of cyclic GMP. In an in vitro kinase assay, SNAP, a NO donor, was shown to directly suppress JNK1 activity, thereby indicating that NO is a direct modulator of JNK1. Moreover, the NO-mediated suppression of JNK1 was demonstrated to be redox-sensitive and dependent on the cysteine-116 in JNK1. Finally, according to the results of an immunohistochemical study using rat striatal neurons, we were able to determine that nNOS-expressing neurons evidenced significantly reduced JNK1 activation. Collectively, these data suggest that JNK1 is regulated by nNOS-mediated NO production in neurons, via a thiol-redox-sensitive mechanism.
Collapse
Affiliation(s)
- Hee-Sae Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Yongbong-dong, Buk-ku, Gwangju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
54
|
Zhao R, Xiang N, Domann FE, Zhong W. Expression of p53 enhances selenite-induced superoxide production and apoptosis in human prostate cancer cells. Cancer Res 2006; 66:2296-304. [PMID: 16489034 PMCID: PMC1435866 DOI: 10.1158/0008-5472.can-05-2216] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although the anticancer effects of selenium have been shown in clinical, preclinical, and laboratory studies, the underlying mechanism(s) remains unclear. Our previous study showed that sodium selenite induced LNCaP human prostate cancer cell apoptosis in association with production of reactive oxygen species, alteration of cell redox state, and mitochondrial damage. In the present study, we showed that selenite-induced apoptosis was superoxide mediated and p53 dependent via mitochondrial pathways. In addition, we also showed that superoxide production by selenite was p53 dependent. Our study showed that wild-type p53-expressing LNCaP cells were more sensitive to selenite-induced apoptosis than p53-null PC3 cells. Selenite treatment resulted in high levels of superoxide production in LNCaP cells but only low levels in PC3 cells. LNCaP cells also showed sequential increases in levels of phosphorylated p53 (serine 15), total p53, Bax, and p21(Waf1) proteins following selenite treatment. The effects of selenite were suppressed by pretreatment with a synthetic superoxide dismutase mimic or by knockdown of p53 via RNA interference. LNCaP cells treated with selenite also showed p53 translocation to mitochondria, cytochrome c release into the cytosol, and activation of caspase-9. On the other hand, restoration of wild-type p53 expression in PC3 cells increased cellular sensitivity to selenite and resulted in increased superoxide production, caspase-9 activation, and apoptosis following selenite treatment. These results suggest that selenite induces apoptosis by producing superoxide to activate p53 and to induce p53 mitochondrial translocation. Activation of p53 in turn synergistically enhances superoxide production and apoptosis induced by selenite.
Collapse
Key Words
- apoptosis
- mitochondria
- p53 tumor suppressor
- prostate cancer
- redox regulation
- selenite
- superoxide
- cuznsod, copper and zinc-containing superoxide dismutase
- h2ax, phosphorylated histone h2ax (serine 139)
- gsh, reduced glutathione
- gssg, glutathione disulfide
- gpx, glutathione peroxidase
- mnsod, manganese-containing superoxide dismutase
- mntmpyp, manganese (iii) tetrakis (n-methyl-2-pyridyl) porphyrin
- moi, multiplicity of infectivity
- mtt, 3-[4,5-dimethyl-2-thiazolyl]-2, 5-diphenyl-2 tetrazolium bromide
- rlu, relative light unit
- ros, reactive oxygen species
- sel, selenite
- sirna, small interfering rna
- sod, superoxide dismutase
Collapse
Affiliation(s)
- Rui Zhao
- The Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison, WI 53792
| | - Nong Xiang
- The Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison, WI 53792
| | - Frederick E. Domann
- Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA 52242 and
| | - Weixiong Zhong
- The Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison, WI 53792
- Pathology and Laboratory Medicine Service, William S. Middleton Veterans Memorial Hospital, Madison, WI 53705
| |
Collapse
|
55
|
Chung YW, Kim TS, Lee SY, Lee SH, Choi Y, Kim N, Min BM, Jeong DW, Kim IY. Selenite-induced apoptosis of osteoclasts mediated by the mitochondrial pathway. Toxicol Lett 2006; 160:143-50. [PMID: 16111838 DOI: 10.1016/j.toxlet.2005.06.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 06/27/2005] [Indexed: 01/26/2023]
Abstract
The possible effects of sodium selenite on mature osteoclasts were investigated. Incubation of osteoclast-like cells differentiated from RAW 264.7 cells with sodium selenite induced apoptosis as revealed by morphological changes, internucleosomal DNA fragmentation, and activation of caspase-3. Selenite also induced generation of the superoxide anion and reduced the number of free thiol groups in the osteoclast-like cells, suggestive of a shift to a more oxidizing intracellular environment. In addition, selenite induced protein aggregation by thiol cross-linking, loss of the mitochondrial membrane potential, and cytochrome c release in mitochondria isolated from the osteoclast-like cells. Finally, selenite-induced DNA fragmentation in osteoclasts was inhibited both by cyclosporin A, a blocker of the mitochondrial permeability transition pore, and by DEVD-CHO, a cell-permeable inhibitor of caspase-3. These results thus suggest that selenite induces apoptosis mediated by the mitochondrial pathway in mature osteoclasts.
Collapse
Affiliation(s)
- Youn Wook Chung
- Laboratory of Cellular and Molecular Biochemistry, School of Life Sciences and Biotechnology, Korea University, Seoul
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Borges VC, Rocha JBT, Nogueira CW. Effect of diphenyl diselenide, diphenyl ditelluride and ebselen on cerebral Na+, K+-ATPase activity in rats. Toxicology 2005; 215:191-7. [PMID: 16095793 DOI: 10.1016/j.tox.2005.07.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Revised: 07/04/2005] [Accepted: 07/06/2005] [Indexed: 10/25/2022]
Abstract
In the present study, we investigated the in vitro effect of diphenyl ditelluride, diphenyl diselenide and ebselen on Na(+), K(+)-ATPase activity of rat brain. The results demonstrated that all compounds significantly inhibited (in the muM range) Na(+), K(+)-ATPase activity. Diphenyl ditelluride, at low concentrations, provoked an increase in Na(+), K(+)-ATPase activity. Dithiothreitol (DTT), at 3mM, protected the inhibition caused by diphenyl ditelluride, diphenyl diselenide and ebselen in Na(+), K(+)-ATPase activity. Post-incubation of diphenyl diselenide-treated homogenate with DTT completely recovered enzyme activity. DTT was able to recover the enzyme inhibition induced by 20muM of diphenyl ditelluride, but was partially able to recover inhibition induced by high concentrations of organotellurium compound. Conversely, DTT did not recover ebselen-induced Na(+), K(+)-ATPase inhibition. The mechanism of inhibition by diphenyl diselenide, diphenyl ditelluride and ebselen in Na(+), K(+)-ATPase activity revealed: decreased maximal velocity and K(m). Cerebral Na(+), K(+)-ATPase is a potential molecular target for the toxic effect of organochalcogens and the inhibition may occur through a change in the crucial thiol groups of this enzyme.
Collapse
Affiliation(s)
- V C Borges
- Departamento de Quimica, Centro de Ciencias Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | | | | |
Collapse
|
57
|
Kim JW, Kim MJ, Kim KJ, Yun HJ, Chae JS, Hwang SG, Chang TS, Park HS, Lee KW, Han PL, Cho SG, Kim TW, Choi EJ. Notch interferes with the scaffold function of JNK-interacting protein 1 to inhibit the JNK signaling pathway. Proc Natl Acad Sci U S A 2005; 102:14308-13. [PMID: 16179393 PMCID: PMC1242280 DOI: 10.1073/pnas.0501600102] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The transmembrane protein Notch is cleaved by gamma-secretase to yield an active form, Notch intracellular domain (Notch-IC), in response to the binding of ligands, such as Jagged. Notch-IC contributes to the regulation of a variety of cellular events, including cell fate determination during embryonic development as well as cell growth, differentiation, and survival. We now show that Notch1-IC suppresses the scaffold activity of c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP1) in the JNK signaling pathway. Notch1-IC physically associated with the JNK binding domain of JIP1 and thereby interfered with the interaction between JIP1 and JNK. JIP1 mediated the activation of JNK1 induced by glucose deprivation in mouse embryonic fibroblasts, and ectopic expression of Notch1-IC inhibited JNK activation and apoptosis triggered by glucose deprivation. Taken together, these findings suggest that Notch1-IC negatively regulates the JNK pathway by disrupting the scaffold function of JIP1.
Collapse
Affiliation(s)
- Jin Woo Kim
- National Creative Research Initiative Center for Cell Death, School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
McEligot AJ, Yang S, Meyskens FL. REDOX REGULATION BY INTRINSIC SPECIES AND EXTRINSIC NUTRIENTS IN NORMAL AND CANCER CELLS. Annu Rev Nutr 2005; 25:261-95. [PMID: 16011468 DOI: 10.1146/annurev.nutr.25.050304.092633] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells in multicellular organisms are exposed to both endogenous oxidative stresses generated metabolically and to oxidative stresses that originate from neighboring cells and from other tissues. To protect themselves from oxidative stress, cells are equipped with reducing buffer systems (glutathione/GSH and thioredoxin/thioredoxin reductase) and have developed several enzymatic mechanisms against oxidants that include catalase, superoxide dismutase, and glutathione peroxidase. Other major extrinsic defenses (from the diet) include ascorbic acid, beta-carotene and other carotenoids, and selenium. Recent evidence indicates that in addition to their antioxidant function, several of these redox species and systems are involved in regulation of biological processes, including cellular signaling, transcription factor activity, and apoptosis in normal and cancer cells. The survival and overall well-being of the cell is dependent upon the balance between the activity and the intracellular levels of these antioxidants as well as their interaction with various regulatory factors, including Ref-1, nuclear factor-kappaB, and activating protein-1.
Collapse
Affiliation(s)
- Archana Jaiswal McEligot
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California 92868, USA.
| | | | | |
Collapse
|
59
|
Qu W, Liu J, Fuquay R, Shimoda R, Sakurai T, Saavedra JE, Keefer LK, Waalkes MP. The nitric oxide prodrug, V-PYRRO/NO, protects against cadmium toxicity and apoptosis at the cellular level. Nitric Oxide 2005; 12:114-20. [PMID: 15740985 DOI: 10.1016/j.niox.2005.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Revised: 11/22/2004] [Accepted: 01/25/2005] [Indexed: 01/22/2023]
Abstract
The liver is an important target tissue of cadmium. The compound O2-vinyl 1-(pyrrolidin-1-yl)diazen-1-ium-1,2 diolate (V-PYRRO/NO) is a liver-selective nitric oxide (NO) prodrug that is metabolized by hepatic P450 enzymes to release NO in hepatocytes. In vivo, V-PYRRO/NO can protect against the toxicity of various hepatotoxicants, including cadmium. Since NO is an effective vasodilator, whether this protective effect against cadmium toxicity is at the level of the hepatic vascular system or actually within the liver cells has not been defined. Thus, we studied the effects of V-PYRRO/NO pretreatment on cadmium-induced toxicity and apoptosis in cultured rat liver epithelial (TRL 1215) cells. Cells were pretreated with V-PYRRO/NO at 500 or 1000 microM for up to 24 h, then exposed to cadmium (as CdCl2) for additional 24 h and cytotoxicity was measured. Cadmium was significantly less cytotoxic in V-PYRRO/NO (1000 microM) pretreated cells (LC50=6.1+/-0.6 microM) compared to control cells (LC50=3.5+/-0.4 microM). TRL 1215 cells acted upon the prodrug to release NO, producing nitrite levels in the extracellular media after 24 h of exposure to 500 or 1000 microM V-PYRRO/NO measured at 87.0+/-4.2 and 324+/-14.8 microM, respectively, compared to basal levels of 7.70+/-0.46 microM. V-PYRRO/NO alone produced small increases in metallothionein (MT), a metal-binding protein associated with cadmium tolerance. However, V-PYRRO/NO pretreatment greatly enhanced cadmium induction of MT. V-PYRRO/NO pretreatment also markedly reduced apoptotic cell death induced by cadmium (5 microM), apparently by blocking cadmium-induced activation of the c-Jun N-terminal kinase (JNK) pathway. Thus, the prodrug, V-PYRRO/NO, protects against the adverse effects of cadmium directly within rat liver cells apparently through generation of NO and, at least in part, by facilitation of cadmium-induced MT synthesis.
Collapse
Affiliation(s)
- Wei Qu
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at the National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
60
|
Hu H, Jiang C, Ip C, Rustum YM, Lü J. Methylseleninic acid potentiates apoptosis induced by chemotherapeutic drugs in androgen-independent prostate cancer cells. Clin Cancer Res 2005; 11:2379-88. [PMID: 15788689 DOI: 10.1158/1078-0432.ccr-04-2084] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To test whether and how selenium enhances the apoptosis potency of selected chemotherapeutic drugs in prostate cancer (PCA) cells. EXPERIMENTAL DESIGN DU145 and PC3 human androgen-independent PCA cells were exposed to minimal apoptotic doses of selenium and/or the topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN38), the topoisomerase II inhibitor etoposide or the microtubule inhibitor paclitaxel/taxol. Apoptosis was measured by ELISA for histone-associated DNA fragments, by flow cytometric analysis of sub-G(1) fraction, and by immunoblot analysis of cleaved poly(ADP-ribose)polymerase. Pharmacologic inhibitors were used to manipulate caspases and c-Jun-NH(2)-terminal kinases (JNK). RESULTS The methylselenol precursor methylseleninic acid (MSeA) increased the apoptosis potency of SN38, etoposide, or paclitaxel by several folds higher than the expected sum of the apoptosis induced by MSeA and each drug alone. The combination treatment did not further enhance JNK1/2 phosphorylation that was induced by each drug in DU145 cells. The JNK inhibitor SP600125 substantially decreased the activation of caspases and apoptosis induced by MSeA combination with SN38 or etoposide and completely blocked these events induced by MSeA/paclitaxel. The caspase-8 inhibitor zIETDfmk completely abolished apoptosis and caspase-9 and caspase-3 cleavage, whereas the caspase-9 inhibitor zLEHDfmk significantly decreased caspase-3 cleavage and apoptosis but had no effect on caspase-8 cleavage. None of these caspase inhibitors abolished JNK1/2 phosphorylation. A JNK-independent suppression of survivin by SN38 and etoposide, but not by paclitaxel, was also observed. In contrast to MSeA, selenite did not show any enhancing effect on the apoptosis induced by these drugs. CONCLUSIONS MSeA enhanced apoptosis induced by cancer therapeutic drugs in androgen-independent PCA cells. In DU145 cells, the enhancing effect was primarily through interactions between MSeA and JNK-dependent targets to amplify the caspase-8-initiated activation cascades. The results suggest a novel use of methyl selenium for improving the chemotherapy of PCA.
Collapse
Affiliation(s)
- Hongbo Hu
- Hormel Institute, University of Minnesota, 801 16th Avenue Northeast, Austin, MN 55912, USA
| | | | | | | | | |
Collapse
|
61
|
Carvalho-Filho MA, Ueno M, Hirabara SM, Seabra AB, Carvalheira JBC, de Oliveira MG, Velloso LA, Curi R, Saad MJA. S-nitrosation of the insulin receptor, insulin receptor substrate 1, and protein kinase B/Akt: a novel mechanism of insulin resistance. Diabetes 2005; 54:959-967. [PMID: 15793233 DOI: 10.2337/diabetes.54.4.959] [Citation(s) in RCA: 210] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Evidence demonstrates that exogenous nitric oxide (NO) and the NO produced by inducible nitric oxide synthase (iNOS) can induce insulin resistance in muscle. Here, we investigated whether this insulin resistance could be mediated by S-nitrosation of proteins involved in early steps of the insulin signal transduction pathway. Exogenous NO donated by S-nitrosoglutathione (GSNO) induced in vitro and in vivo S-nitrosation of the insulin receptor beta subunit (IRbeta) and protein kinase B/Akt (Akt) and reduced their kinase activity in muscle. Insulin receptor substrate (IRS)-1 was also rapidly S-nitrosated, and its expression was reduced after chronic GSNO treatment. In two distinct models of insulin resistance associated with enhanced iNOS expression-diet-induced obesity and the ob/ob diabetic mice-we observed enhanced S-nitrosation of IRbeta/IRS-1 and Akt in muscle. Reversal of S-nitrosation of these proteins by reducing iNOS expression yielded an improvement in insulin action in both animal models. Thus, S-nitrosation of proteins involved in insulin signal transduction is a novel molecular mechanism of iNOS-induced insulin resistance.
Collapse
Affiliation(s)
- Marco A Carvalho-Filho
- Department of Internal Medicine, State University of Campinas, UNICAMP, Campinas, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Abstract
Selenium (Se) is involved in the process of male reproduction. Several studies have been carried out to find the mechanism of Se action through identified selenoproteins. Especially selenoenzyme phospholipid glutathione peroxidase (PHGPx, GPx-4) plays a pivotal role in regulating spermatogenesis. However, the action of selenium is best known as an antioxidant which acts through various selenoproteins viz. glutathione peroxidase, thioredoxin reductase and selenoprotein P. Oxidative stress is currently being considered a leading cause of male infertility. Presently, the involvement of redox active transcription factor, AP1 (Activator protein1) in testicular function was studied. AP1 is redox sensitive and also controls cell proliferation. The effects of Se might be mediated through it. Different Se status - deficient, adequate and excess Se - were generated in male Balb/c mice by feeding yeast based selenium deficient diet and deficient diet supplemented with Se as sodium selenite (0.2 and 1 ppm Se), respectively, for a period of 4 and 8 weeks. Se status was checked by measuring the Se levels and glutathione peroxidase (GSH-Px) activity in testis and liver. The reproductive potential of mice was affected at these changed Se levels. Changes in the activity of superoxide dismutase (SOD), levels of reduced glutathione (GSH) and oxidized glutathione (GSSG) were observed indicating increased oxidative stress at both the levels. Further, changes in the mRNA expression of GSH-Px, gamma-glutamylcysteine synthetase gammaGCS) and Mn superoxide dismutase (MnSOD) were observed. Decrease in cjun and cfos mRNA levels were observed at both the Se status (deficient and excess) which might be responsible for decreased germ cell number, differentiation and reduced fertility observed at the altered Se levels.
Collapse
Affiliation(s)
- Sonia Shalini
- Department of Biophysics, Panjab University, Chandigarh 160014, India
| | | |
Collapse
|
63
|
|
64
|
Schweizer U, Bräuer AU, Köhrle J, Nitsch R, Savaskan NE. Selenium and brain function: a poorly recognized liaison. ACTA ACUST UNITED AC 2004; 45:164-78. [PMID: 15210302 DOI: 10.1016/j.brainresrev.2004.03.004] [Citation(s) in RCA: 223] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2004] [Indexed: 01/08/2023]
Abstract
Molecular biology has recently contributed significantly to the recognition of selenium (Se)2 and Se-dependent enzymes as modulators of brain function. Increased oxidative stress has been proposed as a pathomechanism in neurodegenerative diseases including, among others, Parkinson's disease, stroke, and epilepsy. Glutathione peroxidases (GPx), thioredoxin reductases, and one methionine-sulfoxide-reductase are selenium-dependent enzymes involved in antioxidant defense and intracellular redox regulation and modulation. Selenium depletion in animals is associated with decreased activities of Se-dependent enzymes and leads to enhanced cell loss in models of neurodegenerative disease. Genetic inactivation of cellular GPx increases the sensitivity towards neurotoxins and brain ischemia. Conversely, increased GPx activity as a result of increased Se supply or overexpression ameliorates the outcome in the same models of disease. Genetic inactivation of selenoprotein P leads to a marked reduction of brain Se content, which has not been achieved by dietary Se depletion, and to a movement disorder and spontaneous seizures. Here we review the role of Se for the brain under physiological as well as pathophysiological conditions and highlight recent findings which open new vistas on an old essential trace element.
Collapse
Affiliation(s)
- Ulrich Schweizer
- Neurobiology of Selenium, Neuroscience Research Center, Charité, University Medical School, Berlin, Germany
| | | | | | | | | |
Collapse
|
65
|
Abstract
Mitochondria were classically recognized as the organelles that produce the energy required to drive the endergonic processes of cell life, but now they are considered as the most important cellular source of free radicals, as the main target for free radical regulatory and toxic actions, and as the source of signaling molecules that command cell cycle, proliferation and apoptosis. The progress in the knowledge of mitochondrial functions in the last decades is reviewed. The mitochondrial production of the primary free radicals superoxide anion (O(2)(-)) and nitric oxide (NO), as well as of the termination products H(2)O(2) (hydrogen peroxide) and peroxynitrite (ONOO(-)), is described. A network of intramitochondrial antioxidants consisting of the enzymes Mn-superoxide dismutase and glutathione peroxidase and of the reductants NADH(2), ubiquinol and reduced glutathione, is operative in minimizing the potentially harmful effects of O(2)(-), NO, H(2)O(2) and ONOO(-). Nitric oxide and H(2)O(2) participate in cell signaling, through narrow concentration ranges that signal for opposite cellular situations, i.e., proliferation or apoptosis. A mechanism involving mitogen-activated protein kinases is described. The role of mitochondria in apoptosis is well established through the mitochondrion-dependent pathways of cell death, that includes increased NO production, loss of membrane potential, appearance of dysfunctional mitochondria, cytochrome c release and opening of the voltage-dependent anion channel of the outer membrane.
Collapse
Affiliation(s)
- Enrique Cadenas
- Department of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, PSC-622, 1985 Zonal Avenue, Los Angeles, CA 90033, USA.
| |
Collapse
|
66
|
Meuillet E, Stratton S, Prasad Cherukuri D, Goulet AC, Kagey J, Porterfield B, Nelson MA. Chemoprevention of prostate cancer with selenium: an update on current clinical trials and preclinical findings. J Cell Biochem 2004; 91:443-58. [PMID: 14755676 DOI: 10.1002/jcb.10728] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Prostate cancer is the most common cancer diagnosed and the second leading cause of cancer-related deaths in men in the United States. The etiological factors that give rise to prostate cancer are not known. Therefore, it is not possible to develop primary intervention strategies to remove the causative agents from the environment. However, secondary intervention strategies with selenium (Se) compounds and other agents represent a viable option to reduce the morbidity and mortality of prostate cancer. In this review, we discuss ongoing clinical trials. In addition, we discuss preclinical mechanistic studies that provide insights into the biochemical and molecular basis for the anti-carcinogenic activity of both inorganic and organic forms of Se.
Collapse
Affiliation(s)
- Emmanuelle Meuillet
- Department of Molecular and Cellular Biology, Arizona Cancer Center, The University of Arizona College of Medicine, Tucson, Arizona 85724, USA.
| | | | | | | | | | | | | |
Collapse
|
67
|
Chandrasekher G, Sailaja D. Alterations in lens protein tyrosine phosphorylation and phosphatidylinositol 3-kinase signaling during selenite cataract formation. Curr Eye Res 2004; 28:135-44. [PMID: 14972719 DOI: 10.1076/ceyr.28.2.135.26232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE Protein tyrosine phosphorylation is an important event in the cell signal transduction process. Phosphatidylinositol-3 kinase (PI-3K) is an intracellular signal mediator and plays a key role in many cellular functions. In this study we have examined the changes in lens protein tyrosine phosphorylation and its impact on phosphatidylinositol 3-kinase (PI-3K) signaling during selenite cataract development. METHODS Cataract was induced in 10 days old rat pups by a single sub-cutaneous injection of sodium selenite (30 microM/Kg body weight) and lenses were collected at different stages of cataract development. Immunoprecipitation and Western immunoblotting were employed to determine protein tyrosine phosphorylation, PI-3K activity and protein in lens cell extracts. Tyrosine kinase activity in lens membrane preparations was assayed in the presence of a synthetic substrate peptide and [32P]ATP. RESULTS Protein tyrosine phosphorylation in the lens was disrupted before the onset of cataract. A decrease in tyrosine phosphorylation of lens proteins was observed within 2-3 days of selenite injection (pre-cataract stage). The effect was much more prominent with the progression of cataract. The decrease in protein tyrosine phosphorylation correlated with the decrease in tyrosine kinase activity associated with the lens membrane fraction. Stimulation of normal rat lenses in organ culture with insulin and IGF-1 caused an increase in the phosphorylation of proteins, whose tyrosine phosphorylation status appeared to be diminished during cataract development. Insulin and IGF-1 also stimulated rat lens PI-3K activity. While there was no change in total PI-3K activity during the onset of cataract, the activity of PI-3K associated with tyrosine phosphorylated proteins decreased markedly in pre-cataract lenses. Further, the ability of IGF-1 to stimulate PI-3K activity was significantly reduced in lens epithelial cells treated with selenium. CONCLUSIONS These studies show that signaling events involving the protein tyrosine phosphorylation process and activation of PI-3K are altered during selenite cataract formation and implicate defects in signal transduction mechanisms as contributing factors in the development of cataract.
Collapse
Affiliation(s)
- Gudiseva Chandrasekher
- Department of Ophthalmology and Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | |
Collapse
|
68
|
Kim SH, Johnson VJ, Shin TY, Sharma RP. Selenium attenuates lipopolysaccharide-induced oxidative stress responses through modulation of p38 MAPK and NF-kappaB signaling pathways. Exp Biol Med (Maywood) 2004; 229:203-13. [PMID: 14734799 DOI: 10.1177/153537020422900209] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lipopolysaccharide (LPS) produces reactive oxygen species (ROS) and nitric oxide (NO) in macrophages. These molecules are involved in inflammation associated with endotoxic shock. Selenium (Se), a biologically essential trace element, modulates the functions of many regulatory proteins involved in signal transduction and affects a variety of cellular activities, including cell growth and survival. We demonstrate that Se attenuated LPS-induced ROS and NO production in murine macrophage cultures in vitro. This Se-decreased production of NO was demonstrated by decreases in both mRNA and protein expression for inducible NO synthase (iNOS). The preventive effects of Se on iNOS were p38 mitogen-activated protein kinase- and nuclear factor-kappaB-dependent. Se specifically blocked the LPS-induced activation of p38 but not that of c-jun-N-terminal kinase and extracellular signal-regulated kinase; the p38-specific pathway was confirmed using p38 inhibitor SB 203580. These results suggest that the mechanism by which Se may act as an anti-inflammatory agent and that Se may be considered as a possible preventive intervention for endotoxemia, particularly in Se-deficient locations. However, the efficacy and safety of Se need to be further investigated, because long-term intake > 0.4 mg Se/day in adults can produce adverse effects.
Collapse
Affiliation(s)
- Sang Hyun Kim
- Department of Physiology and Pharmacology, College of Veterinary Medicine, The University of Georgia, Athens, 30602, USA
| | | | | | | |
Collapse
|
69
|
Park HS, Yu JW, Cho JH, Kim MS, Huh SH, Ryoo K, Choi EJ. Inhibition of apoptosis signal-regulating kinase 1 by nitric oxide through a thiol redox mechanism. J Biol Chem 2003; 279:7584-90. [PMID: 14668338 DOI: 10.1074/jbc.m304183200] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide is an endogenous thiol-reactive molecule that modulates the functions of many regulatory proteins by a thiol-redox mechanism. NO has now been shown to inhibit the activation of apoptosis signal-regulating kinase 1 (ASK1) in murine fibrosarcoma L929 cells through such a mechanism. Exposure of L929 cells to interferon-gamma resulted in the endogenous production of NO and in inhibition of the activation of ASK1 by hydrogen peroxide. The interferon-gamma-induced inhibition of ASK1 activity was blocked by N(G)-nitro-l-arginine, an inhibitor of NO synthase. Furthermore, the NO donor S-nitro-N-acetyl-dl-penicillamine (SNAP) inhibited ASK1 activity in vitro, and this inhibition was reversed by thiol-reducing agents such as dithiothreitol and beta-mercaptoethanol. SNAP did not inhibit the kinase activities of MKK3, MKK6, or p38 in vitro. The inhibition of ASK1 by interferon-gamma was not changed by 1H- (1,2,4)oxadiazolo[4,3-alpha]quinoxalin-1-one, an inhibitor of guanylyl cyclase nor was it mimicked by 8-bromo-cyclic GMP. Site-directed mutagenesis revealed that replacement of cysteine 869 of ASK1 by serine rendered this protein resistant to the inhibitory effects both of interferon-gamma in intact cells and of SNAP in vitro. Co-immunoprecipitation data showed that NO production inhibited a binding of ASK1, but not ASK1(C869S), to MKK3 or MKK6. Moreover, interferon-gamma induced the S-nitrosylation of endogenous ASK1 in L929 cells. Together, these results suggest that NO mediates the interferon-gamma-induced inhibition of ASK1 in L929 cells through a thiolredox mechanism.
Collapse
Affiliation(s)
- Hee-Sae Park
- National Creative Research Initiative Center for Cell Death and School of Life Science and Biotechnology, Korea University, Seoul, 136-701, Korea
| | | | | | | | | | | | | |
Collapse
|
70
|
Sarker KP, Biswas KK, Rosales JL, Yamaji K, Hashiguchi T, Lee KY, Maruyama I. Ebselen inhibits NO-induced apoptosis of differentiated PC12 cells via inhibition of ASK1-p38 MAPK-p53 and JNK signaling and activation of p44/42 MAPK and Bcl-2. J Neurochem 2003; 87:1345-53. [PMID: 14713291 DOI: 10.1046/j.1471-4159.2003.02096.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Ebselen, a selenium-containing heterocyclic compound, prevents ischemia-induced cell death. However, the molecular mechanism through which ebselen exerts its cytoprotective effect remains to be elucidated. Using sodium nitroprusside (SNP) as a nitric oxide (NO) donor, we show here that ebselen potently inhibits NO-induced apoptosis of differentiated PC12 cells. This was associated with inhibition of NO-induced phosphatidyl Serine exposure, cytochrome c release, and caspase-3 activation by ebselen. Analysis of key apoptotic regulators during NO-induced apoptosis of differentiated PC12 cells showed that ebselen blocks the activation of the apoptosis signaling-regulating kinase 1 (ASK1), and inhibits phosphorylation of p38 mitogen-activated protein kinase (MAPK) and c-jun N-terminal protein kinase (JNK). Moreover, ebselen inhibits NO-induced p53 phosphorylation at Ser15 and c-Jun phosphorylation at Ser63 and Ser73. It appears that inhibition of p38 MAPK and p53 phosphorylation by ebselen occurs via a thiol-redox-dependent mechanism. Interestingly, ebselen also activates p44/42 MAPK, and inhibits the downregulation of the antiapoptotic protein Bcl-2 in SNP-treated PC12 cells. Together, these findings suggest that ebselen protects neuronal cells from NO cytotoxicity by reciprocally regulating the apoptotic and antiapoptotic signaling cascades.
Collapse
Affiliation(s)
- Krishna P Sarker
- Department of Laboratory and Molecular Medicine, Faculty of Medicine, Kagoshima University, Kagoshima-890, Japan.
| | | | | | | | | | | | | |
Collapse
|
71
|
Lee YC, Tang YC, Chen YH, Wong CM, Tsou AP. Selenite-induced survival of HuH7 hepatoma cells involves activation of focal adhesion kinase-phosphatidylinositol 3-kinase-Akt pathway and Rac1. J Biol Chem 2003; 278:39615-24. [PMID: 12896980 DOI: 10.1074/jbc.m304095200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Selenium has been shown to sustain the growth of selected human hepatocellular carcinoma cell lines under serum-free conditions, but the detailed mechanism remained undetermined. In the present study, the molecular mechanism(s) involving sodium selenite (Na2SO3, Se) as a survival agent were determined. Selenite not only protects HuH7 cells from serum deprivation-induced apoptosis, it also supports its long-term growth in sodium selenite (10(-7)m) supplemented serum-free medium. The anti-apoptotic effect correlates with activation of focal adhesion kinase and the phosphatidylinositol 3-kinase (PI3K)-Akt kinase pathway. Using HuH7 cells stably transfected with a constitutively active Akt kinase and PI3K inhibitor LY294002, selenite-induced cell survival was shown to be PI3K-Akt-dependent. Parallel changes included a significant reduction in the intracellular reactive oxygen species content, the reversal of DNA fragmentation, and the suppression of caspase and apoptosis signal-regulating kinase 1 activities. HuH7 cells stably expressing a Rac1 mutant N17 (Rac1N17-HuH7) are refractory to selenite treatment. In these cells selenite supplement neither triggers Akt activation nor supports cell proliferation. Participation of Rac1 activation in this event is supported by the fact that selenite treatment drastically enhanced activation of Rac1. The exact link between selenite treatment, Rac1 activation, and activation of the focal adhesion kinase-PI 3-kinase, however, remains to be characterized. The mitogenic signaling mediated by selenite may involve unconventional growth stimuli including higher glutathione peroxidase 1 activity and higher transcription levels of selenoprotein P. The selenium-HuH7 system we have established thus provides a unique tool that will allow the biological role of selenite in growth regulation of hepatocytes to be studied in detail.
Collapse
Affiliation(s)
- Yu-Chi Lee
- Institute of Genetics, School of Life Sciences, National Yang-Ming University, Shih-Pai, Taipei 112, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
72
|
Nango R, Terada C, Tsukamoto I. Jun N-terminal kinase activation and upregulation of p53 and p21(WAF1/CIP1) in selenite-induced apoptosis of regenerating liver. Eur J Pharmacol 2003; 471:1-8. [PMID: 12809946 DOI: 10.1016/s0014-2999(03)01764-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To investigate apoptosis induced by selenite in hepatocytes in vivo, rats received a single injection of sodium selenite immediately after partial hepatectomy. Characteristic DNA fragmentation in gel electrophoresis and in situ end-labeling and the increase in caspase-3 activity were observed at 4 h after partial hepatectomy with selenite injection. The activation of Jun N-terminal kinase (JNK) was observed as early as 15 min and increased to about 10-fold the maximal level of the control at 1 and 2 h after partial hepatectomy in selenite-injected rats, while a transient increase was observed at 1 h in the control. Western blot analysis revealed that the c-Jun and the phosphorylated c-Jun protein markedly increased after 30 min and reached a maximal level at 1 and 2 h after partial hepatectomy with selenite injection, although c-Jun and a faint band of the phosphorylated c-Jun were observed after 1 h in the control. The levels of c-jun mRNA and c-Fos protein and mRNA in selenite-injected rats also increased more than in the control. The rise in the p53 protein level after partial hepatectomy with selenite injection was followed by the upregulation of p21(WAF1/CIP1) mRNA and protein expression. These results suggested that selenite induced apoptosis accompanied by the activation of caspase-3 and JNK and the upregulation of c-jun, c-fos, p53 and p21(WAF1/CIP1) at the early stage of liver regeneration.
Collapse
Affiliation(s)
- Rieko Nango
- Department of Food Science and Nutrition, Nara Women's University, Nara 630, Japan
| | | | | |
Collapse
|
73
|
Schroeter H, Boyd CS, Ahmed R, Spencer JPE, Duncan RF, Rice-Evans C, Cadenas E. c-Jun N-terminal kinase (JNK)-mediated modulation of brain mitochondria function: new target proteins for JNK signalling in mitochondrion-dependent apoptosis. Biochem J 2003; 372:359-69. [PMID: 12614194 PMCID: PMC1223409 DOI: 10.1042/bj20030201] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2003] [Revised: 02/24/2003] [Accepted: 03/04/2003] [Indexed: 01/05/2023]
Abstract
The molecular mechanisms underlying the initiation and control of the release of cytochrome c during mitochondrion-dependent apoptosis are thought to involve the phosphorylation of mitochondrial Bcl-2 and Bcl-x(L). Although the c-Jun N-terminal kinase (JNK) has been proposed to mediate the phosphorylation of Bcl-2/Bcl-x(L) the mechanisms linking the modification of these proteins and the release of cytochrome c remain to be elucidated. This study was aimed at establishing interdependency between JNK signalling and mitochondrial apoptosis. Using an experimental model consisting of isolated, bioenergetically competent rat brain mitochondria, these studies show that (i) JNK catalysed the phosphorylation of Bcl-2 and Bcl-x(L) as well as other mitochondrial proteins, as shown by two-dimensional isoelectric focusing/SDS/PAGE; (ii) JNK-induced cytochrome c release, in a process independent of the permeability transition of the inner mitochondrial membrane (imPT) and insensitive to cyclosporin A; (iii) JNK mediated a partial collapse of the mitochondrial inner-membrane potential (Deltapsim) in an imPT- and cyclosporin A-independent manner; and (iv) JNK was unable to induce imPT/swelling and did not act as a co-inducer, but as an inhibitor of Ca-induced imPT. The results are discussed with regard to the functional link between the Deltapsim and factors influencing the permeability transition of the inner and outer mitochondrial membranes. Taken together, JNK-dependent phosphorylation of mitochondrial proteins including, but not limited to, Bcl-2/Bcl-x(L) may represent a potential of the modulation of mitochondrial function during apoptosis.
Collapse
Affiliation(s)
- Hagen Schroeter
- Department of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles 90089-9121, USA
| | | | | | | | | | | | | |
Collapse
|
74
|
Preston TJ, Woodgett JR, Singh G. JNK1 activity lowers the cellular production of H2O2 and modulates the growth arrest response to scavenging of H2O2 by catalase. Exp Cell Res 2003; 285:146-58. [PMID: 12681294 DOI: 10.1016/s0014-4827(03)00015-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hydrogen peroxide (H(2)O(2)) can interact with intracellular signaling pathways to regulate cell behavior. The c-Jun NH(2)-terminal kinase 1 (JNK1) signal, involved in diverse aspects of cellular functioning, is implicated as a cell sensor of redox stress. The growth-inhibitory effect of both high-level H(2)O(2) and H(2)O(2)-scavenging catalase treatments is accompanied by increased JNK1 activity. To investigate the role of this response in growth regulation, the JNK1 signal was increased by the introduction of ectopic HA-JNK1. HA-JNK1 expression correlated with increases in basal c-Jun phosphorylation in a dose-dependent manner. Transient expression of HA-JNK1 potentiated cell growth arrest by catalase; however, with stable expression a degree of resistance to this response was observed. Resistance was accompanied by a lowered endogenous production of H(2)O(2). Transient HA-JNK1 expression also reduced H(2)O(2) generation, and this effect was reversed by the JNK inhibitor SP600125. These results indicate that the JNK1 stress response contributes to growth inhibition by catalase treatment via inhibition of cellular H(2)O(2) production. Stable amplification of the JNK1 pathway leads to cellular adaptation to its signal, resulting in a diminished reliance upon H(2)O(2) for efficient growth.
Collapse
|
75
|
Cheng WH, Zheng X, Quimby FR, Roneker CA, Lei XG. Low levels of glutathione peroxidase 1 activity in selenium-deficient mouse liver affect c-Jun N-terminal kinase activation and p53 phosphorylation on Ser-15 in pro-oxidant-induced aponecrosis. Biochem J 2003; 370:927-34. [PMID: 12492400 PMCID: PMC1223242 DOI: 10.1042/bj20021870] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2002] [Revised: 12/18/2002] [Accepted: 12/19/2002] [Indexed: 11/17/2022]
Abstract
Low levels of hepatic selenium (Se)-dependent glutathione peroxidase 1 (GPX1) activity have been shown to protect against oxidative liver injury in Se-deficient mice. The objective of the present study was to determine if the GPX1 protection was associated with phosphorylations of c-Jun N-terminal kinase (JNK) and p53 on Ser-15, two key signalling events in oxidative-stress-mediated cell death. Both Se-deficient GPX1 knockout (GPX1(-/-)) and wild-type (WT) mice ( n =64) were pretreated with an intraperitoneal injection of Se (as sodium selenite, 50 microg/kg body weight) 6 h before an intraperitoneal injection of paraquat (12.5 mg/kg). Liver aponecrosis, a mixed form of cell death sharing apoptosis and necrosis, was induced by paraquat in both groups of mice. However, its appearance was remarkably delayed and the severity was decreased by the repletion of hepatic GPX1 activity to <4% of the normal level by the Se injection in the WT mice, compared with that in the GPX1(-/-) mice. Consistently, the WT mice had lower levels of hepatic phospho-JNK, p53 and phospho-p53 (Ser-15) when compared with the GPX1(-/-) mice at 1-10 h after paraquat injection. Incubating liver homogenates with antibodies raised against JNK or phospho-JNK resulted in co-immunoprecipitation of phospho-p53 (Ser-15), and the amounts of the precipitated phospho-p53 were greater in the GPX1(-/-) mice when compared with that in the WT mice. The co-precipitated complex by the anti-phospho-JNK antibody was capable of phosphorylating intrinsic or extrinsic p53 on Ser-15. In conclusion, phospho-JNK may catalyse phosphorylation of p53 on Ser-15 in Se-deficient mouse liver under moderate oxidative stress, and attenuation of that cascade by low levels of GPX1 activity is associated with its protection against the pro-oxidant-induced liver aponecrosis.
Collapse
Affiliation(s)
- Wen-Hsing Cheng
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | |
Collapse
|
76
|
Larabee JL, Hocker JR, Hanas RJ, Kahn FM, Hanas JS. Inhibition of zinc finger protein-DNA interactions by sodium selenite. Biochem Pharmacol 2002; 64:1757-65. [PMID: 12445865 DOI: 10.1016/s0006-2952(02)01414-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sodium selenite and sodium selenate were analyzed for their ability to alter the DNA binding mechanisms of the Cys(2)His(2) zinc finger proteins, transcription factor IIIA (TFIIIA) and Sp1. TFIIIA is a positive regulator of 5S ribosomal RNA synthesis, and Sp1 is involved in cell proliferation and invasiveness. As assayed by DNase I protection, the interaction of the DNA binding domain of TFIIIA with the 5S ribosomal gene was inhibited by 25 microM selenite ions but not by 250 microM selenate ions. Selenite inhibition kinetics of TFIIIA progressed to completion in about 5 min. Preincubation of free TFIIIA with selenite resulted in DNA binding inhibition, whereas preincubation of a TFIIIA/5S RNA complex with selenite did not. Since 5S RNA binds to the TFIIIA DNA binding domain, this result is consistent with an inhibition mechanism via selenite binding to that region of this protein. Inhibition was not readily reversible and occurred in the presence of an excess of beta-mercaptoethanol; elevated amounts of dithiothreitol mitigated the inhibitory effect. Significantly less selenite (2.5-5 microM) inhibited the specific DNA binding of transcription factor Sp1 to the simian virus 40 (SV40) early promoter/enhancer. The selenite inhibition kinetics of Sp1 were fast, going to completion in about 1 min. SV40 DNA binding by the non-zinc finger transcription factor AP-2 was not inhibited by selenite. Inhibition of Cys(2)His(2) zinc finger proteins by micromolar amounts of selenite points to additional mechanisms for selenite-induced diminution of cell growth and anticancer activity.
Collapse
Affiliation(s)
- Jason L Larabee
- Department of Biochemistry and Molecular Biology, University of Oklahoma College of Medicine, 800 Research Parkway, Room 448, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
77
|
Haddad JJ. Pharmaco-redox regulation of cytokine-related pathways: from receptor signaling to pharmacogenomics. Free Radic Biol Med 2002; 33:907-26. [PMID: 12361802 DOI: 10.1016/s0891-5849(02)00985-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines represent a multi-diverse family of polypeptide regulators; they are relatively low molecular weight (< 30 kDa), pharmacologically active proteins that are secreted by one cell for the purpose of altering either its own functions (autocrine effect) or those of adjacent cells (paracrine effect). Cytokines are small, nonenzymatic glycoproteins whose actions are both diverse and overlapping (specificity/redundancy) and may affect diverse and overlapping target cell populations. In many instances, individual cytokines have multiple biological activities. Different cytokines can also have the same activity, which provides for functional redundancy (network) within the inflammatory and immune systems. As biological cofactors that are released by specific cells, cytokines have specific effects on cell-cell interaction, communication, and behavior of other cells. As a result, it is infrequent that loss or neutralization of one cytokine will markedly interfere with either of these systems. The biological effect of one cytokine is often modified or augmented by another. Because an interdigitating, redundant network of cytokines is involved in the production of most biological effects, both under physiologic and pathologic conditions, it usually requires more than a single defect in the network to alter drastically the outcome of the process. This fact, therefore, may have crucial significance in the development of therapeutic strategies for biopharmacologic intervention in cytokine-mediated inflammatory processes and infections.
Collapse
Affiliation(s)
- John J Haddad
- Severinghaus-Radiometer Research Labs, Molecular Neuroscience Research Division, Dept of Anesthesia and Perioperative Care, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0542, USA.
| |
Collapse
|
78
|
Jeong DW, Yoo MH, Kim TS, Kim JH, Kim IY. Protection of mice from allergen-induced asthma by selenite: prevention of eosinophil infiltration by inhibition of NF-kappa B activation. J Biol Chem 2002; 277:17871-6. [PMID: 11897787 DOI: 10.1074/jbc.m200808200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The potential anti-inflammatory effect of sodium selenite in a mouse model of asthma was investigated. Selenite was injected into the peritoneum of allergen (ovalbumin)-sensitized mice before allergen challenge. Ovalbumin challenge resulted in activation of the transcription factor NF-kappaB and an increase in the expression of cell adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and E-selectin, which are encoded by NF-kappaB-dependent genes) in lung tissue as well as in the recruitment of eosinophils to lung airways. These effects of ovalbumin challenge were all inhibited by pretreatment of mice with selenite. Selenite administration also increased the activity of selenium-dependent glutathione peroxidase in lung tissue. Furthermore, supplementation of A549 human airway epithelial cell cultures with selenite increased glutathione peroxidase activity as well as inhibited both the generation of hydrogen peroxide and the activation of NF-kappaB induced by tumor necrosis factor alpha in these cells. Selenite also reversed in vitro the activation of NF-kappaB induced by this cytokine in intact A549 cells. These results suggest that selenite regulates the activity of NF-kappaB by increasing the activity of glutathione peroxidase, thereby removing potential activators of NF-kappaB, and possibly also by direct oxidation of critical sulfhydryl groups of this transcription factor. These effects of selenite likely underlie its anti-inflammatory action in asthma.
Collapse
Affiliation(s)
- Dae-Won Jeong
- Graduate School of Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | |
Collapse
|
79
|
Gupta N, Porter TD. Inhibition of human squalene monooxygenase by selenium compounds. J Biochem Mol Toxicol 2002; 16:18-23. [PMID: 11857773 DOI: 10.1002/jbt.10014] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Selenosis in animals is characterized by a variety of neurological abnormalities, but the chemical species of selenium and the molecular targets that mediate this neurotoxicity are unknown. We have previously shown that selenite is a potent inhibitor of squalene monooxygenase, the second enzyme in the committed pathway for cholesterol biosynthesis; inhibition of this enzyme by dimethyltellurium leads to a peripheral demyelinating neuropathy similar to that seen in selenosis. To evaluate the role methylation plays in selenium toxicity, we examined the ability of three methylselenium compounds, methylselenol, dimethylselenide, and trimethylselenonium iodide, to inhibit purified recombinant human squalene monooxygenase. IC(50) values for methylselenol (95 microM) and dimethylselenide (680 microM) were greater than that previously obtained for selenite (37 microM), and inhibition by trimethylselenonium iodide was evident only at concentrations above 3 mM. Inhibition by methylselenol as well as by selenite was slow and irreversible, suggestive of covalent binding to the enzyme, and thiol-containing compounds could prevent and reverse this inhibition, indicating that these compounds were reacting with sulfhydryl groups on the protein. Monothiols such as glutathione and beta-mercaptoethanol provided better protection than did dithiols, suggesting that these selenium compounds bind to only one of the two proposed vicinal cysteines on squalene monooxygenase. Unexpectedly, the inhibition by selenite was significantly enhanced by dithiols, indicating that a more toxic species, possibly selenide, was formed in the presence of these dithiol reductants.
Collapse
Affiliation(s)
- Nisha Gupta
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0082, USA
| | | |
Collapse
|
80
|
Abstract
The essential role of selenium (Se) in nutrition is well established. The elucidation of the mechanisms by which selenium regulates the cell cycle can lead to a better understanding of the nature of selenium's essentiality and its role in disease prevention. In this study, the effects of selenium deficiency or adequacy (0.25 micromol/L selenite or selenomethionine) on HL-60 cell cycle progression were examined in serum-free media. Selenium was critical for promotion of HL-60 cell growth. Cell-cycle analysis revealed that selenium deficiency caused a decrease in G1 phase cells that corresponded to an increase in G2 and sub-G1 phase cells. Gene array analysis suggested that c-Myc, cyclin C, proliferating cell nuclear antigen, cyclin-dependent kinase (cdk)1, cdk2, cdk4, cyclin B and cyclin D2 mRNA levels were lower in selenium-deficient cells than in the cells supplemented with 0.25 micromol/L selenomethionine. The decrease in the c-Myc mRNA level in selenium-deficient cells was confirmed by reverse transcription-polymerase chain reaction analysis. Furthermore, the phosphorylation state of total cellular protein was higher (57%) in selenium-supplemented cells than in selenium-deficient cells. Collectively, these results suggest a novel role for selenium at 0.25 micromol/L in up-regulation of the expression of numerous cell cycle-related genes and total cellular phosphorylated proteins in HL-60 cells in serum-free culture media. This leads to the promotion of cell cycle progression, particularly G2/M transition and/or the reduction of apoptosis, primarily in G1 cells. These observations may have additional implications for understanding the nature of selenium's essentiality.
Collapse
Affiliation(s)
- Huawei Zeng
- U.S. Department of Agriculture, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58202-9034, USA.
| |
Collapse
|
81
|
McKenzie RC, Arthur JR, Beckett GJ. Selenium and the regulation of cell signaling, growth, and survival: molecular and mechanistic aspects. Antioxid Redox Signal 2002; 4:339-51. [PMID: 12006185 DOI: 10.1089/152308602753666398] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In the past 30 years, it has been recognized that dietary selenium (Se) is essential for the normal function of many of the systems of the body. Furthermore, low Se intake can have deleterious effects on several aspects of human and animal health. The importance of Se is characterized in its role as a constituent of several key antioxidant and redox enzyme families. Most of the effects of Se are probably mediated by selenoproteins, which have the micronutrient covalently incorporated into the protein. The purpose of this review is to examine basic mechanisms by which Se regulates cell growth, gene transcription, cell signaling, and cell death. We start with the historical background to Se. The synthesis and function of selenoproteins are described, followed by details of the dietary sources of Se and Se status in different parts of the world, together with the clinical effects of Se deficiency and toxicity. We consider some aspects of the molecular mechanisms by which Se modulates cell growth, intracellular signaling, and gene transcription.
Collapse
Affiliation(s)
- Roderick C McKenzie
- Department of Medical and Radiological Sciences, University of Edinburgh, Edinburgh, EH3 9YW, Scotland, UK.
| | | | | |
Collapse
|
82
|
Bébien M, Lagniel G, Garin J, Touati D, Verméglio A, Labarre J. Involvement of superoxide dismutases in the response of Escherichia coli to selenium oxides. J Bacteriol 2002; 184:1556-64. [PMID: 11872706 PMCID: PMC134873 DOI: 10.1128/jb.184.6.1556-1564.2002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Selenium can provoke contrasting effects on living organisms. It is an essential trace element, and low concentrations have beneficial effects, such as the reduction of the incidence of cancer. However, higher concentrations of selenium salts can be toxic and mutagenic. The bases for both toxicity and protection are not clearly understood. To provide insights into these mechanisms, we analyzed the proteomic response of Escherichia coli cells to selenate and selenite treatment under aerobic conditions. We identified 23 proteins induced by both oxides and ca. 20 proteins specifically induced by each oxide. A striking result was the selenite induction of 8 enzymes with antioxidant properties, particularly the manganese and iron superoxide dismutases (SodA and SodB). The selenium inductions of sodA and sodB were controlled by the transcriptional regulators SoxRS and Fur, respectively. Strains with decreased superoxide dismutase activities were severely impaired in selenium oxide tolerance. Pretreatment with a sublethal selenite concentration triggered an adaptive response dependent upon SoxRS, conferring increased selenite tolerance. Altogether, our data indicate that superoxide dismutase activity is essential for the cellular defense against selenium salts, suggesting that superoxide production is a major mechanism of selenium toxicity under aerobic conditions.
Collapse
Affiliation(s)
- Magali Bébien
- CEN/Cadarache-DSV-DEVM Laboratoire de Bioénergétique Cellulaire, Univ-Méditérranée CEA 1000, 13108 Saint Paul-Lez-Durance Cedex, France
| | | | | | | | | | | |
Collapse
|
83
|
Utsugi M, Dobashi K, Koga Y, Shimizu Y, Ishizuka T, Iizuka K, Hamuro J, Nakazawa T, Mori M. Glutathione redox regulates lipopolysaccharide‐induced IL‐12 production through p38 mitogen‐activated protein kinase activation in human monocytes: role of glutathione redox in IFN‐γ priming of IL‐12 production. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.2.339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Mitsuyoshi Utsugi
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Kunio Dobashi
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Yasuhiko Koga
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Yasuo Shimizu
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Tamotsu Ishizuka
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Kunihiko Iizuka
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| | - Junji Hamuro
- Basic Research Laboratories, Ajinomoto Co., Kawasaki, Japan
| | - Tsugio Nakazawa
- Faculty of Health Sciences, Gunma University, Maebashi, Japan; and
| | - Masatomo Mori
- First Department of Internal Medicine, Faculty of Medicine, School of Medicine, and Kawasaki, Japan
| |
Collapse
|
84
|
Park HS, Kim MS, Huh SH, Park J, Chung J, Kang SS, Choi EJ. Akt (protein kinase B) negatively regulates SEK1 by means of protein phosphorylation. J Biol Chem 2002; 277:2573-8. [PMID: 11707464 DOI: 10.1074/jbc.m110299200] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The protein serine-threonine kinase Akt mediates cell survival signaling initiated by various growth-promoting factors such as insulin. Here we report that SEK1 is a target of Akt in intact cells. Insulin inhibited the anisomycin-induced stimulation of both endogenous SEK1 and its substrate c-Jun N-terminal kinase (JNK), but not that of the upstream kinase MEKK1, in 293T cells. The inhibitory action of insulin on SEK1 or JNK1 activation was prevented by the phosphatidylinositol 3-kinase inhibitor LY294002. Expression of a constitutively active form of Akt also inhibited both SEK1 and JNK1 activation, but not that of MEKK1, in transfected 293T cells. Co-immunoprecipitation analysis revealed that endogenous Akt physically interacted with endogenous SEK1 in cells and that this interaction was promoted by insulin. In vitro and in vivo (32)P labeling indicated that Akt phosphorylated SEK1 on serine 78. The SEK1 mutant SEK1(S78A) was resistant to Akt-induced inhibition. Finally, activated Akt inhibited SEK1-mediated apoptosis, and this effect of Akt was prevented by overexpression of SEK(S78A). Taken together, these results suggest that Akt suppresses stress-activated signaling by targeting SEK1.
Collapse
Affiliation(s)
- Hee-Sae Park
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | | | | | |
Collapse
|
85
|
Kim JY, Park HS, Kang SI, Choi EJ, Kim IY. Redox regulation of cytosolic glycerol-3-phosphate dehydrogenase: Cys(102) is the target of the redox control and essential for the catalytic activity. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1569:67-74. [PMID: 11853959 DOI: 10.1016/s0304-4165(01)00236-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cytosolic glycerol-3-phosphate dehydrogenase (cG3PDH) occupies the branch point between the glycolytic pathway and triglyceride biosynthesis. However, the regulatory mechanism of the cG3PDH activity has remained obscure. Here we report that cG3PDH is efficiently inhibited by modification of the thiol group through a redox mechanism. In this study, we found that sodium selenite and nitric oxide (NO) donors such as S-nitroso-N-acetylpenicillamine and 3-morpholinosydnonimine inhibited cG3PDH activity, and that similar effects could be achieved with selenium metabolites such as selenocysteine and selenomethionine. Furthermore, we found that reducing agents, such as dithiothreitol and beta-mercaptoethanol, restored the cG3PDH activity suppressed by selenite and NO both in vitro and in cultured cells. Buthionine sulfoximine depleted levels of both reduced glutathione and the oxidized form but had no effect on the suppression of cG3PDH activity by selenite in cultured cells. Moreover, thiol-reactive agents, such as N-ethylmaleimide and o-iodosobenzoic acid, blocked the enzyme activity of cG3PDH through the modification of redox-sensitive cysteine residues in cG3PDH. The inhibitor of NO synthase, L-N(G)-nitro-arginine, restored the cG3PDH activity inhibited by NO in cultured cells, whereas the inhibitor of guanylyl cyclase, 1H-[1,2,4] oxadiazole[4,3-alpha] quinoxalin-1-one (ODQ), has no effect. NO directly inhibits cG3PDH activity not via a cGMP-dependent mechanism. Finally, using site-directed mutagenesis, we found that Cys(102) of cG3PDH was sensitive to both selenite and NO. From the results, we suggest that cG3PDH is a target of cellular redox regulation.
Collapse
Affiliation(s)
- Ji-Young Kim
- Laboratory of Cellular and Molecular Biochemistry, Graduate School of Biotechnology, Korea University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
86
|
Yoon SO, Kim MM, Park SJ, Kim D, Chung J, Chung AS. Selenite suppresses hydrogen peroxide-induced cell apoptosis through inhibition of ASK1/JNK and activation of PI3-K/Akt pathways. FASEB J 2002; 16:111-3. [PMID: 11709494 DOI: 10.1096/fj.01-0398fje] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The relationship between selenium and signal molecules has not been well elucidated. It was found that physiological concentration of selenite, 3 microM, reduced ASK1 activity and induced PI3-kinase (PI3-K)/Akt pathways in HT1080 cells. Duration of these signal molecules by selenite was much longer than that by growth factors and other stresses. The longer duration time of these signal molecules may be important to maintain normal functions against stresses. Selenite increased cell proliferation through up-regulation of Bcl-2 expression, mitochondrial membrane potential, adenosine triphosphate (ATP) generation, and glucose uptake mediated by PI3-K pathway. High concentration of H2O2 increased an apoptotic signal molecule, ASK1, which resulted in Bcl-2 down-regulation, membrane potential disruption, decreased ATP and glucose uptake, and activation of caspases. However, an antiapoptotic signal molecule, Akt, was activated also by H2O2, but duration of its activation was much shorter. Selenite blocked apoptosis induced by H2O2, which was related to blocking ASK1 and further stimulating PI3-kinase/Akt activities. Selenite blocked mitochondrial membrane potential disruption by 400 mM H2O2. Selenite also blocked caspase-9 and -3 activities and apoptosis induced by 500 microM H2O2, even after mitochondrial membrane potential disruption. These observations demonstrate that selenite increases cell proliferation and maintains cell survival by activating the antiapoptotic signal and blocking the apoptotic signal.
Collapse
Affiliation(s)
- Sang-Oh Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Taejon 305-701, South Korea
| | | | | | | | | | | |
Collapse
|
87
|
Goss GG, Jiang L, Vandorpe DH, Kieller D, Chernova MN, Robertson M, Alper SL. Role of JNK in hypertonic activation of Cl(-)-dependent Na(+)/H(+) exchange in Xenopus oocytes. Am J Physiol Cell Physiol 2001; 281:C1978-90. [PMID: 11698257 DOI: 10.1152/ajpcell.2001.281.6.c1978] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the course of studying the hypertonicity-activated ion transporters in Xenopus oocytes, we found that activation of endogenous oocyte Na(+)/H(+) exchange activity (xoNHE) by hypertonic shrinkage required Cl(-), with an EC(50) for bath [Cl(-)] of approximately 3mM. This requirement for chloride was not supported by several nonhalide anions and was not shared by xoNHE activated by acid loading. Hypertonicity-activated xoNHE exhibited an unusual rank order of inhibitory potency among amiloride derivatives and was blocked by Cl(-) transport inhibitors. Chelation of intracellular Ca(2+) by injection of EGTA blocked hypertonic activation of xoNHE, although many inhibitors of Ca(2+)-related signaling pathways were without inhibitory effect. Hypertonicity activated oocyte extracellular signal-regulated kinase 1/2 (ERK1/2), but inhibitors of neither ERK1/2 nor p38 prevented hypertonic activation of xoNHE. However, hypertonicity also stimulated a Cl(-)-dependent increase in c-Jun NH(2)-terminal kinase (JNK) activity. Inhibition of JNK activity prevented hypertonic activation of xoNHE but not activation by acid loading. We conclude that hypertonic activation of Na(+)/H(+) exchange in Xenopus oocytes requires Cl(-) and is mediated by activation of JNK.
Collapse
Affiliation(s)
- G G Goss
- Department of Biological Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E9
| | | | | | | | | | | | | |
Collapse
|
88
|
Fu Y, Sies H, Lei XG. Opposite roles of selenium-dependent glutathione peroxidase-1 in superoxide generator diquat- and peroxynitrite-induced apoptosis and signaling. J Biol Chem 2001; 276:43004-9. [PMID: 11562367 DOI: 10.1074/jbc.m106946200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oxidative injuries including apoptosis can be induced by reactive oxygen species (ROS) and reactive nitrogen species (RNS) in aerobic metabolism. We determined impacts of a selenium-dependent glutathione peroxidase-1 (GPX1) on apoptosis induced by diquat (DQ), a ROS (superoxide) generator, and peroxynitrite (PN), a potent RNS. Hepatocytes were isolated from GPX1 knockout (GPX1-/-) or wild-type (WT) mice, and treated with 0.5 mm DQ or 0.1-0.8 mm PN for up to 12 h. Loss of cell viability, high levels of apoptotic cells, and severe DNA fragmentation were produced by DQ in only GPX1-/- cells and by PN in only WT cells. These two groups of cells shared similar cytochrome c release, caspase-3 activation, and p21(WAF1/CIP1) cleavage. Higher levels of protein nitration were induced by PN in WT than GPX1-/- cells. Much less and/or slower cellular GSH depletion was caused by DQ or PN in GPX1-/- than in WT cells, and corresponding GSSG accumulation occurred only in the latter. In conclusion, it is most striking that, although GPX1 protects against apoptosis induced by superoxide-generator DQ, the enzyme actually promotes apoptosis induced by PN in murine hepatocytes. Indeed, GSH is a physiological substrate for GPX1 in coping with ROS in these cells.
Collapse
Affiliation(s)
- Y Fu
- Department of Animal Science, Cornell University, Ithaca, New York 14853, USA
| | | | | |
Collapse
|
89
|
LaRochelle O, Gagné V, Charron J, Soh JW, Séguin C. Phosphorylation is involved in the activation of metal-regulatory transcription factor 1 in response to metal ions. J Biol Chem 2001; 276:41879-88. [PMID: 11551972 DOI: 10.1074/jbc.m108313200] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have studied the role of phosphorylation in the activation of metal-regulatory transcription factor-1 (MTF-1) and metallothionein (MT) gene expression. We showed that MTF-1 is phosphorylated in vivo and that zinc stimulates MTF-1 phosphorylation 2-4-fold. Several kinase inhibitors were used to examine the possible involvement of kinase cascades in the activation of MTF-1. Metal-induced MT gene expression was abrogated by protein kinase C (PKC), c-Jun N-terminal kinase (JNK), phosphoinositide 3-kinase, and tyrosine-specific protein kinases inhibitors, as assayed by Northern analysis and by cotransfection experiments using a metal regulatory element-luciferase reporter plasmid. The extracellular signal-activated protein kinase and the p38 kinase cascades did not appear to be essential for the activation of MT gene transcription by metals. By using dominant-negative mutants of PKC, JNK, mitogen-activated kinase kinase 4 (MKK4), and MKK7, we provide further evidence supporting a role for PKC and JNK in the activation of MTF-1 in response to metals. Notably, increased MTF-1 DNA binding in response to zinc and MTF-1 nuclear localization was not inhibited in cells preincubated with the different kinase inhibitors despite strong inhibition of MTF-1-mediated gene expression. This suggests that phosphorylation is essential for MTF-1 transactivation function. We hypothesize that metal-induced phosphorylation of MTF-1 is one of the primary events leading to increased MTF-1 activity. Thus, metal ions such as cadmium could activate MTF-1 and induce MT gene expression by stimulating one or several kinases in the MTF-1 signal transduction pathway.
Collapse
Affiliation(s)
- O LaRochelle
- Centre de Recherche en Cancérologie de l'Université Laval, CHUQ, Hôtel-Dieu de Québec, Québec, Québec G1R 2J6, Canada
| | | | | | | | | |
Collapse
|
90
|
Affiliation(s)
- H Schindler
- Institute of Clinical Microbiology, Immunology and Hygiene, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany.
| | | |
Collapse
|
91
|
Yoon SO, Kim MM, Chung AS. Inhibitory effect of selenite on invasion of HT1080 tumor cells. J Biol Chem 2001; 276:20085-92. [PMID: 11274215 DOI: 10.1074/jbc.m101143200] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Selenium, an essential biological trace element, has been shown to reduce and prevent the incidence of cancer. Our previous studies have shown that selenite is involved in the chemoprevention of cancer and induction of apoptosis of cancer cells. In this study, we demonstrate that selenite also inhibits the invasion of tumor cells. Cancer cell invasion requires coordinated processes, such as changes in cell-cell and cell-matrix adhesion, degradation of the extracellular matrix, and cell migration. We found that selenite inhibited invasion of HT1080 human fibrosarcoma cells. Adhesion of HT1080 cells to the collagen matrix was also inhibited by treatment with selenite, but cell-cell interaction and cell motility were not affected by selenite. Moreover, selenite reduced expression of matrix metalloproteinase-2 and -9 and urokinase-type plasminogen activator, which are involved in matrix degradation, but increased a tissue inhibitor of metalloproteinase-1. This inhibitory effect of selenite on the protease expressions was mediated by the suppression of transcription factors, NF-kappaB and AP-1. However, selenate showed no remarkable effect on all the steps of cancer cell invasion.
Collapse
Affiliation(s)
- S O Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Taejon 305-701, South Korea
| | | | | |
Collapse
|
92
|
Kim JW, Chang TS, Lee JE, Huh SH, Yeon SW, Yang WS, Joe CO, Mook-Jung I, Tanzi RE, Kim TW, Choi EJ. Negative regulation of the SAPK/JNK signaling pathway by presenilin 1. J Cell Biol 2001; 153:457-63. [PMID: 11331298 PMCID: PMC2190568 DOI: 10.1083/jcb.153.3.457] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Presenilin 1 (PS1) plays a pivotal role in Notch signaling and the intracellular metabolism of the amyloid beta-protein. To understand intracellular signaling events downstream of PS1, we investigated in this study the action of PS1 on mitogen-activated protein kinase pathways. Overexpressed PS1 suppressed the stress-induced stimulation of stress-activated protein kinase (SAPK)/c-Jun NH(2)-terminal kinase (JNK) in human embryonic kidney 293 cells. Interestingly, two functionally inactive PS1 mutants, PS1(D257A) and PS1(D385A), failed to inhibit UV-stimulated SAPK/JNK. Furthermore, H(2)O(2-) or UV-stimulated SAPK activity was higher in mouse embryonic fibroblast (MEF) cells from PS1-null mice than in MEF cells from PS(+/+) mice. MEF(PS1(-/-)) cells were more sensitive to the H(2)O(2)-induced apoptosis than MEF(PS1(+/+)) cells. Ectopic expression of PS1 in MEF(PS1(-/-)) cells suppressed H(2)O(2)-stimulated SAPK/JNK activity and apoptotic cell death. Together, our data suggest that PS1 inhibits the stress-activated signaling by suppressing the SAPK/JNK pathway.
Collapse
Affiliation(s)
- Jin Woo Kim
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| | - Tong-Shin Chang
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| | - Ji Eun Lee
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| | - Sung-Ho Huh
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| | - Seung Woo Yeon
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Kyongki-do, Korea
| | - Wan Seok Yang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Taejon, 305-701, Korea
| | - Cheol O. Joe
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Taejon, 305-701, Korea
| | - Inhee Mook-Jung
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Kyongki-do, Korea
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Tae-Wan Kim
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Eui-Ju Choi
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| |
Collapse
|
93
|
Abstract
Accumulated evidence from prospective studies, intervention trials and studies on animal models of cancer have suggested a strong inverse correlation between selenium intake and cancer incidence. Several putative mechanisms have been suggested to mediate the chemopreventive activities of selenium: of these, the inhibition of cellular proliferation and the induction of apoptosis are particularly attractive. The mitogen activated protein kinase (MAPK) pathways are known to be important regulators of cell death and our recent work has focused on the involvement of these pathways in selenium-induced apoptosis in primary cultures of oral cancers and corresponding normal mucosa derived from biopsy material. Using this system, the oral carcinoma cells were found to have enhanced sensitivity to apoptosis when treated with certain selenium compounds compared to normal oral mucosa. Induction of Fas ligand was associated with selenium-induced apoptosis. Signal transduction studies suggests that selenium induces several changes in the MAPK signalling pathways but functional intervention/inhibitor studies indicate that activation of the JNK pathway seems to be most important.
Collapse
Affiliation(s)
- A Ghose
- CRC Beatson Laboratories, The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | | | | |
Collapse
|
94
|
Park HS, Huh SH, Kim MS, Lee SH, Choi EJ. Nitric oxide negatively regulates c-Jun N-terminal kinase/stress-activated protein kinase by means of S-nitrosylation. Proc Natl Acad Sci U S A 2000; 97:14382-7. [PMID: 11121042 PMCID: PMC18927 DOI: 10.1073/pnas.97.26.14382] [Citation(s) in RCA: 201] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
NO, produced from l-arginine in a reaction catalyzed by NO synthase, is an endogenous free radical with multiple functions in mammalian cells. Here, we demonstrate that endogenously produced NO can suppress c-Jun N-terminal kinase (JNK) activation in intact cells. Treatment of BV-2 murine microglial cells with IFN-gamma induced endogenous NO production, concomitantly suppressing JNK1 activation. Similarly, IFN-gamma induced suppression of JNK1 activation in RAW264.7 murine macrophage cells and rat alveolar macrophages. The IFN-gamma-induced suppression of JNK1 activation in BV-2, RAW264.7, or rat alveolar macrophage cells was completely prevented by N(G)-nitro-l-arginine, a NO synthase inhibitor. Interestingly, the IFN-gamma-induced suppression of JNK1 activation was not affected by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, an inhibitor of guanylyl cyclase. 8-Bromo-cGMP, a membrane-permeant analogue of cGMP, did not change JNK1 activation in intact cells either. In contrast, S-nitro-N-acetyl-dl-penicillamine (SNAP), a NO donor, inhibited JNK1 activity in vitro. Furthermore, a thiol reducing agent, DTT, reversed not only the in vitro inhibition of JNK1 activity by SNAP but also the in vivo suppression of JNK1 activity by IFN-gamma. Substitution of serine for cysteine-116 in JNK1 abolished the inhibitory effect of IFN-gamma or SNAP on JNK1 activity in vivo or in vitro, respectively. Moreover, IFN-gamma enhanced endogenous S-nitrosylation of JNK1 in RAW264.7 cells. Collectively, our data suggest that endogenous NO mediates the IFN-gamma-induced suppression of JNK1 activation in macrophage cells by means of a thiol-redox mechanism.
Collapse
Affiliation(s)
- H S Park
- National Creative Research Initiative Center for Cell Death, Graduate School of Biotechnology, Korea University, Seoul, 136-701, Korea
| | | | | | | | | |
Collapse
|
95
|
Hall JP, Merithew E, Davis RJ. c-Jun N-terminal kinase (JNK) repression during the inflammatory response? Just say NO. Proc Natl Acad Sci U S A 2000; 97:14022-4. [PMID: 11121010 PMCID: PMC34087 DOI: 10.1073/pnas.97.26.14022] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- J P Hall
- Howard Hughes Medical Institute, Program in Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | |
Collapse
|