1
|
D'Souza V, RJ M, Shetty M, A. V, Chakraborthy A, B. MK, A. VS, Badanthadka M. Protein Malnutrition in BALB/c Mice: An Experimental Model Resembling Clinical Scenario. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2022. [DOI: 10.1055/s-0042-1758526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
Objectives The study aims to develop a stable malnourished experimental mice model resembling the human population for future experimental studies.
Materials and Methodology At weaning, female BALB/c mice are separated into two groups: one receiving a low protein diet (LPD, 10% protein) and the other receiving a commercially available normal pellet diet (ND, 18% protein). Model development and stability were assessed using body mass index (BMI), biochemical parameters such as glucose, aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, total protein, albumin, total cholesterol, calcium, and phosphorus using serum samples at the 12th and 15th weeks of the study, antioxidant assay, and liver histopathology observation. Antioxidant assay and histopathology observation using liver tissue sample excised after euthanasia.
Results LPD mice are categorized under grade I malnutrition based on the body weight change with respect to ND as per the principles of Gomez's classification of malnutrition. A significant long-term decrease in BMI of the malnourished group indicates the development of the stable malnourished model. Elevated serum enzyme levels in the 15th week and decreased antioxidant activity suggest liver injury and oxidative stress at the cellular level in the malnourished group. Histopathology alterations in the liver tissue further strengthen these observations reported in the human population of malnutrition.
Conclusion This study confirms the development of a stable malnourished experimental model using a LPD (10% protein). This model may be used to study the role of malnutrition in the pathophysiology of any disease, drug action, and its kinetics in the future.
Collapse
Affiliation(s)
- Vinitha D'Souza
- Department of Nitte University Centre for Animal Research and Experimentation (NUCARE), NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Paneer campus, Deralakatte, Mangaluru, Karnataka, India
| | - Madhura RJ
- Department of Nitte University Centre for Animal Research and Experimentation (NUCARE), NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Paneer campus, Deralakatte, Mangaluru, Karnataka, India
| | - Meghashree Shetty
- Department of Nitte University Centre for Animal Research and Experimentation (NUCARE), NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Paneer campus, Deralakatte, Mangaluru, Karnataka, India
| | - Varsha A.
- Department of Nitte University Centre for Animal Research and Experimentation (NUCARE), NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Paneer campus, Deralakatte, Mangaluru, Karnataka, India
| | - Anirban Chakraborthy
- Division of Molecular Genetics and Cancer, Nitte University Centre for Science Education and Research (NUCSER), Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - Mohana Kumar B.
- Nitte University Centre for Stem Cell Research & Regenerative Medicine (NUCSReM), K. S. Hegde Medical Academy (KSHEMA), Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - Veena Shetty A.
- Department of Microbiology, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - Murali Badanthadka
- Department of Nitte University Centre for Animal Research and Experimentation (NUCARE), NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Paneer campus, Deralakatte, Mangaluru, Karnataka, India
| |
Collapse
|
2
|
Cho IJ, Kim D, Kim EO, Jegal KH, Kim JK, Park SM, Zhao R, Ki SH, Kim SC, Ku SK. Cystine and Methionine Deficiency Promotes Ferroptosis by Inducing B-Cell Translocation Gene 1. Antioxidants (Basel) 2021; 10:antiox10101543. [PMID: 34679678 PMCID: PMC8532826 DOI: 10.3390/antiox10101543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a type of programmed necrosis triggered by iron-dependent lipid peroxidation. We investigated the role of B-cell translocation gene 1 (BTG1) in cystine and methionine deficiency (CST/Met (−))-mediated cell death. CST/Met (−) depleted reduced and oxidized glutathione in hepatocyte-derived cells, increased prostaglandin-endoperoxide synthase 2 expression, and promoted reactive oxygen species accumulation and lipid peroxidation, as well as necrotic cell death. CST/Met (−)-mediated cell death and lipid peroxidation was specifically inhibited by pretreatment with ferroptosis inhibitors. In parallel with cell death, CST/Met (−) blocked global protein translation and increased the expression of genes associated with the integrated stress response. Moreover, CST/Met (−) significantly induced BTG1 expression. Using a BTG1 promoter-harboring reporter gene and siRNA, activating transcription factor 4 (ATF4) was identified as an essential transcription factor for CST/Met (−)-mediated BTG1 induction. Although knockout of BTG1 in human HAP1 cells did not affect the accumulation of reactive oxygen species induced by CST/Met (−), BTG1 knockout significantly decreased the induction of genes associated with the integrated stress response, and reduced lipid peroxidation and cell death in response to CST/Met (−). The results demonstrate that CST/Met (−) induces ferroptosis by activating ATF4-dependent BTG1 induction.
Collapse
Affiliation(s)
- Il-Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
| | - Doyeon Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
| | - Eun-Ok Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
| | - Kyung-Hwan Jegal
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
- Digital Health Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Jae-Kwang Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
| | - Sang-Mi Park
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
| | - Rongjie Zhao
- Department of Psychopharmacology, Qiqihar Medical University, Qiqihar 161006, China;
| | - Sung-Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61452, Korea;
| | - Sang-Chan Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
- Correspondence: (S.-C.K.); (S.-K.K.); Tel.: +82-53-819-1862 (S.-C.K.); +82-53-819-1549 (S.-K.K.)
| | - Sae-Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea; (I.-J.C.); (D.K.); (E.-O.K.); (K.-H.J.); (J.-K.K.); (S.-M.P.)
- Correspondence: (S.-C.K.); (S.-K.K.); Tel.: +82-53-819-1862 (S.-C.K.); +82-53-819-1549 (S.-K.K.)
| |
Collapse
|
3
|
Effect of Dietary Methionine Deficiency Followed by a Re-Feeding Phase on the Hepatic Antioxidant Activities of Lambs. Animals (Basel) 2020; 11:ani11010007. [PMID: 33374518 PMCID: PMC7822206 DOI: 10.3390/ani11010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Our objective was to investigate the effect of methionine restriction and resuming supply on liver antioxidant response in lambs. The concentrations of methionine and its metabolites and the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2), a redox sensitive factor, were detected after methionine restriction treatment for 50 days and methionine supply recovery for 29 days. The expression of glutathione (GSH) S-transferase (GST), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) were characterized at the level of transcription and translation. Methionine restriction can directly change the content of methionine and its metabolites in plasma and liver, and affect the redox state of lambs by activating the Nrf2 signaling pathway. Liver tissue can adapt to oxidative environment by upregulating the expression of antioxidant enzymes such as GSH-Px and SOD. Moreover, it was found that there was a lag effect in the recovery of metabolism after methionine supplementation.
Collapse
|
4
|
Chowdhury MMR, Park J, Afrin F, Ko YG, Kim CL, Lee SS, Kim SW. Transcriptome profiling of in vitro-matured oocytes from a korean native cow (hanwoo) after cysteamine supplementation. Anim Biotechnol 2020; 32:401-412. [PMID: 31900040 DOI: 10.1080/10495398.2019.1706545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study elucidated the molecular markers that decrease oocyte quality during in vitro culture, restricting optimal developmental potential. Here, we evaluated the transcriptomic differences between cysteamine-treated and non-treated bovine cumulus oocyte complexes (COCs) after 22 h of co-culture in the maturation media using RNA sequencing. In total, 39,014 transcripts were sequenced between cysteamine-treated and non-treated mature COCs. We evaluated the relative expression of 21,472 genes, with 59 genes showing differential expression between the two COC groups. The cysteamine-treated group had 36 up-regulated gene transcripts and 23 down-regulated gene transcripts. Moreover, gene ontology (GO) enrichment analysis revealed that multiple biological processes were significantly enriched after cysteamine supplementation. Differentially expressed genes appeared to maintain normal oocyte physiology, regulation of apoptosis, differentiation, ossification or bone formation, cardiac and muscle physiology, hormonal secretion, and membrane construction for further embryonic development. In conclusion, cysteamine affects the mRNA level of COCs during oocyte maturation by upregulating potential molecular markers and downregulating genes that affect further embryonic development.
Collapse
Affiliation(s)
- M M R Chowdhury
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea.,Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Dhaka, Bangladesh
| | - Joonghoon Park
- Department of International Agricultural Technology, Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Fahmida Afrin
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Yeoung-Gyu Ko
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Chan-Lan Kim
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Sung Soo Lee
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Sung Woo Kim
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| |
Collapse
|
5
|
Caballero VJ, Mendieta JR, Lombardo D, Saceda M, Ferragut JA, Conde RD, Giudici AM. Liver damage and caspase-dependent apoptosis is related to protein malnutrition in mice: effect of methionine. Acta Histochem 2015; 117:126-35. [PMID: 25575574 DOI: 10.1016/j.acthis.2014.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/28/2014] [Accepted: 11/29/2014] [Indexed: 01/18/2023]
Abstract
This study aimed to determine whether the effects on the mouse liver caused by three periods of feeding a protein-free diet for 5 days followed by a normal complete diet for 5 days (3PFD-CD) are prevented by a constant methionine supply (3PFD+Met-CD). The expressions of carbonic anhydrase III (CAIII), fatty acid synthase (FAS), glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and glutathione S-transferase P1 (GSTP1) were assessed by proteomics and reverse transcriptase-polymerase chain reactions. The liver redox status was examined by measuring the activities of superoxide dismutase (SOD) and catalase (CAT), as well as protein carbonylation. Because oxidative stress can result in apoptosis, the activity and content of caspase-3, as well as the x-linked inhibitor of the apoptosis protein (XIAP) and mitochondrial caspase-independent apoptosis inducing factor (AIF) contents were assessed. In addition, the liver histomorphology was examined. Compared to the controls fed a normal complete diet throughout, feeding with 3PFD-CD increased the FAS content, decreased the CAIII content, decreased both the SOD and CAT activities, and increased protein carbonylation. It also activated caspase-3, decreased the XIAP content, decreased the AIF content, increased the number of GSTP1-positive foci and caspase-3-positive cells, and caused fatty livers. Conversely, the changes were lessened to varying degrees in mice fed 3PFD+Met-CD. The present results indicate that a regular Met supply lessens the biochemical changes, damage, and caspase-dependent apoptosis provoked by recurrent dietary amino acid deprivation in the mouse liver.
Collapse
Affiliation(s)
- Verónica J Caballero
- Biological Research Institute, Faculty of Natural Sciences, National University of Mar del Plata - CONICET, CC 1245, CP 7600 Mar del Plata, Argentina
| | - Julieta R Mendieta
- Biological Research Institute, Faculty of Natural Sciences, National University of Mar del Plata - CONICET, CC 1245, CP 7600 Mar del Plata, Argentina
| | - Daniel Lombardo
- Institute of Research and Technology in Animal Reproduction (INITRA), Faculty of Veterinary Science. University of Buenos Aires, Av. Chorroarín 280, C1427CWO Buenos Aires, Argentina
| | - Miguel Saceda
- Institute of Molecular and Cellular Biology, University Miguel Hernandez, Building Torregaitán, Avda de la Universidad s/n, 03202, Elche, Spain
| | - José Antonio Ferragut
- Institute of Molecular and Cellular Biology, University Miguel Hernandez, Building Torregaitán, Avda de la Universidad s/n, 03202, Elche, Spain
| | - Rubén D Conde
- Biological Research Institute, Faculty of Natural Sciences, National University of Mar del Plata - CONICET, CC 1245, CP 7600 Mar del Plata, Argentina
| | - Ana M Giudici
- Biological Research Institute, Faculty of Natural Sciences, National University of Mar del Plata - CONICET, CC 1245, CP 7600 Mar del Plata, Argentina.
| |
Collapse
|
6
|
Gum SI, Cho MK. Differential hepatic GSTA2 expression of arylalkyl isothiocyanates in vivo and in vitro: The molecular mechanism of gene induction by phenethyl isothiocyanate. Mol Nutr Food Res 2013; 57:2223-32. [DOI: 10.1002/mnfr.201300259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/26/2013] [Accepted: 06/30/2013] [Indexed: 01/07/2023]
Affiliation(s)
- Sang Il Gum
- Department of Pharmacology; College of Oriental Medicine; Dongguk University; Kyungju Korea
| | - Min Kyung Cho
- Department of Pharmacology; College of Oriental Medicine; Dongguk University; Kyungju Korea
| |
Collapse
|
7
|
Liver disease and malnutrition. Best Pract Res Clin Gastroenterol 2013; 27:619-29. [PMID: 24090946 DOI: 10.1016/j.bpg.2013.06.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/01/2013] [Accepted: 06/12/2013] [Indexed: 01/31/2023]
Abstract
Patients with hepatic disorders are exceptionally vulnerable to developing malnutrition because of the key role played by the liver in regulating the nutritional state and the energy balance. Moreover, the presence of chronic liver disorders could reduce the appetite and thus influence the nutrient intake. Poor nutritional status has been shown in various patient groups with hepatic disorders, and particularly in patients with alcoholic cirrhosis who are at high nutritional risk. It is well established that malnourished patients with liver diseases generally have a higher risk of developing adverse clinical outcomes and increased healthcare costs. Nutrition screening with the Subjective Global Assessment and anthropometric measurements are an important first step in the early identification of malnutrition and initiates the whole nutrition care process. It is therefore important for appropriate nutrition policies and protocols to be implemented so that all patients with chronic liver diseases are monitored closely from a nutritional standpoint. Early and evidence-based nutritional interventions are eagerly needed to minimize the nutritional decline associated with chronic liver disorders and ultimately improve the prognosis of such patients. This review includes a comprehensive analysis of methods to identify malnutrition in patients with chronic liver diseases as well as the extent and impact of the malnutrition problem in selected patient populations.
Collapse
|
8
|
Lin AH, Chen HW, Liu CT, Tsai CW, Lii CK. Activation of Nrf2 is required for up-regulation of the π class of glutathione S-transferase in rat primary hepatocytes with L-methionine starvation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:6537-6545. [PMID: 22676582 DOI: 10.1021/jf301567m] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Numerous genes expression is regulated in response to amino acid shortage, which helps organisms adapt to amino acid limitation. The expression of the π class of glutathione (GSH) S-transferase (GSTP), a highly inducible phase II detoxification enzyme, is regulated mainly by activates activating protein 1 (AP-1) binding to the enhancer I of GSTP (GPEI). Here we show the critical role of nuclear factor erythroid-2-related factor 2 (Nrf2) in up-regulating GSTP gene transcription. Primary rat hepatocytes were cultured in a methionine-restricted medium, and immunoblotting and RT-PCR analyses showed that methionine restriction time-dependently increased GSTP protein and mRNA expression over a 48 h period. Nrf2 translocation to the nucleus, nuclear proteins binding to GPEI, and antioxidant response element (ARE) luciferase reporter activity were increased by methionine restriction as well as by l-buthionine sulfoximine (BSO), a GSH synthesis inhibitor. Transfection with Nrf2 siRNA knocked down Nrf2 expression and reversed the methionine-induced GSTP expression and GPEI binding activity. Chromatin immunoprecipitation assay confirmed the binding of Nrf2 to the GPEI. Phosphorylation of extracellular signal-regulated kinase 2 (ERK2) was increased in methionine-restricted and BSO-treated cells. ERK2 siRNA abolished methionine restriction-induced Nrf2 nuclear translocation, GPEI binding activity, ARE-luciferase reporter activity, and GSTP expression. Our results suggest that the up-regulation of GSTP gene transcription in response to methionine restriction likely occurs via the ERK-Nrf2-GPEI signaling pathway.
Collapse
Affiliation(s)
- Ai-Hsuan Lin
- School of Nutrition, Chung Shan Medical University, Taichung 402, Taiwan
| | | | | | | | | |
Collapse
|
9
|
Caballero VJ, Mendieta JR, Giudici AM, Crupkin AC, Barbeito CG, Ronchi VP, Chisari AN, Conde RD. Alternation between dietary protein depletion and normal feeding cause liver damage in mouse. J Physiol Biochem 2010; 67:43-52. [PMID: 20878513 DOI: 10.1007/s13105-010-0047-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 09/15/2010] [Indexed: 01/12/2023]
Abstract
The effect of frequent protein malnutrition on liver function has not been intensively examined. Thus, the effects of alternating 5 days of a protein and amino acid-free diet followed by 5 days of a complete diet repeated three times (3 PFD-CD) on female mouse liver were examined. The expression of carbonic anhydrase III (CAIII), fatty acid synthase (FAS), glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and glutathione S-transferase P1 (GSTP1) in liver were assessed by proteomics, reverse transcriptase-polymerase chain reaction and Northern blotting. The activities of liver GSTs, glutathione reductase (GR) and catalase (CAT), as well as serum glutamic-oxaloacetic transaminase (SGOT) and glutamic-pyruvic transaminase (SGPT) were also tested. Additionally, oxidative damage was examined by measuring of protein carbonylation and lipid peroxidation. Liver histology was examined by light and electron microscopy. Compared with control mice, 3 PFD-CD increased the content of FAS protein (+90%) and FAS mRNA (+30%), while the levels of CAIII and CAIII mRNAs were decreased (-48% and -64%, respectively). In addition, 3 PFD-CD did not significantly change the content of GSTP1 but produced an increase in its mRNA level (+20%), while it decreased the activities of both CAT (-66%) and GSTs (-26%). After 3 PFD-CD, liver protein carbonylation and lipid peroxidation were increased by +55% and +95%, respectively. In serum, 3 PFD-CD increased the activities of both SGOT (+30%) and SGPT (+61%). In addition, 3 PFD-CD showed a histological pattern characteristic of hepatic damage. All together, these data suggest that frequent dietary amino acid deprivation causes hepatic metabolic and ultrastructural changes in a fashion similar to precancerous or cancerous conditions.
Collapse
Affiliation(s)
- Veronica J Caballero
- Instituto de Investigaciones Biológicas, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata-CONICET, Funes 3250 4º Nivel, CP 7600, Mar del Plata, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Oxidative stress in mouse liver caused by dietary amino acid deprivation: protective effect of methionine. J Physiol Biochem 2010; 66:93-103. [PMID: 20577846 DOI: 10.1007/s13105-010-0014-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 03/30/2010] [Indexed: 01/24/2023]
Abstract
The aim of this work was to evaluate the effects of a diet depleted of amino acids (protein-free diet, or PFD), as well as the supplementation with methionine (PFD+Met), on the antioxidant status of the female mouse liver. With this purpose, cytosolic protein spots from two-dimensional non-equilibrium pH gel electrophoresis were identified by several procedures, such as mass spectrometry, Western blot, gel matching and enzymatic activity. PFD decreased the contents of catalase (CAT), peroxiredoxin I (Prx-I), and glutathione peroxidase (GPx) by 67%, 37% and 45%, respectively. Gene expression analyses showed that PFD caused a decrease in CAT (-20%) and GPx (-30%) mRNA levels but did not change that of Prx-I. It was also found that, when compared to a normal diet, PFD increased the liver contents of both reactive oxygen species (+50%) and oxidized protein (+88%) and decreased that of glutathione (-45%). Supplementation of PFD with Met prevented these latter effects to varying degrees, whereas CAT, Prx-I and GPx mRNA levels resulted unmodified. Present results suggest that dietary amino acid deprivation deranges the liver antioxidant defences, and this can be, in part, overcome by supplementation with Met.
Collapse
|
11
|
Tsai CW, Lin AH, Wang TS, Liu KL, Chen HW, Lii CK. Methionine restriction up-regulates the expression of the pi class of glutathione S
-transferase partially via
the extracellular signal-regulated kinase-activator protein-1 signaling pathway initiated by glutathione depletion. Mol Nutr Food Res 2009; 54:841-50. [DOI: 10.1002/mnfr.200900083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
12
|
Galhardi F, Mesquita K, Monserrat J, Barros D. Effect of silymarin on biochemical parameters of oxidative stress in aged and young rat brain. Food Chem Toxicol 2009; 47:2655-60. [DOI: 10.1016/j.fct.2009.07.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 07/13/2009] [Accepted: 07/24/2009] [Indexed: 12/14/2022]
|
13
|
Sidhu P, Garg ML, Dhawan DK. Protective Effects of Zinc on Oxidative Stress Enzymes in Liver of Protein-Deficient Rats. Drug Chem Toxicol 2008; 28:211-30. [PMID: 15865262 DOI: 10.1081/dct-52551] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Persons afflicted with protein malnutrition are generally deficient in a variety of essential micronutrients like zinc, copper, iron, and selenium, which in turn affects number of metabolic processes in the body. To evaluate the protective effects of zinc on the enzymes involved in oxidative stress induced in liver of protein-deficient rats, the current study was designed. Zinc sulfate at a dose level of 227 mg/L zinc in drinking water was administered to female Sprague-Dawley normal control as well as protein-deficient rats for a total duration of 8 weeks. The effects of zinc treatment in conditions of protein deficiency were studied on rat liver antioxidant enzymes, which included catalase, glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD), glutathione reduced (GSH), and glutathione-S-transferase (GST). Protein deficiency in normal rats resulted in a significant increase in hepatic activities of catalase, glutathione peroxidase, glutathione reductase, and glutathione-S-transferase and the levels of lipid peroxidation. A significant inhibition in the levels of reduced glutathione and the enzyme activity of superoxide dismutase has been observed after protein deficiency in normal rats. Interestingly, Zn treatment to protein-deficient animals lowered already raised activity catalase, glutathione peroxidase, and glutathione-S-transferase and levels of lipid peroxidation to significant levels when compared to protein-deficient animals. Also, Zn treatment to the protein-deficient animals resulted in a significant elevation in the levels of GSH and SOD activity as compared to their respective controls, thereby indicating its effectiveness in regulating their levels in adverse conditions. It has also been observed that concentrations of zinc, copper, iron, and selenium were found to be decreased significantly in protein-deficient animals. However, the levels of these elements came back to within normal limits when zinc was administrated to protein-deficient rats. This study concludes that zinc has the potential to regulate the activities of oxidative stress enzymes as well as essential hepatic elements.
Collapse
Affiliation(s)
- Pardeep Sidhu
- Institute of Physiology and Experimental Pathophysiology, Friedrich-Alexander University, Erlangen, Germany
| | | | | |
Collapse
|
14
|
Expression of cytokine mRNA in lymphocytes of malnourished children. J Clin Immunol 2008; 28:593-9. [PMID: 18496743 DOI: 10.1007/s10875-008-9204-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 04/11/2008] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Protein-calorie malnutrition represents a significant worldwide health problem and is associated with an increased risk for infections. The purpose of this study was to evaluate possible changes in type 1/type 2 responses balance in malnourished children. RESULTS The data obtained in the present study showed that the expression levels of tumor necrosis factor-alpha, interleukin (IL)-4, and IL-10 were more highly, in contrast IL-2, gamma interferon, and IL-6 genes were expressed less in all groups of malnourished children compared with the well-nourished infected children. It is important to indicate that the data collected in the present work agree with the results obtained by different authors, who showed differences in the production of cytokines in malnourished children. CONCLUSION In conclusion, the results suggest that alterations in the balance of type 1/type 2 immune responses exist in malnourished children, and this could be the reason that the immunological system of the malnourished children is incapable of eradicating infections.
Collapse
|
15
|
Casalino E, Calzaretti G, Landriscina M, Sblano C, Fabiano A, Landriscina C. The Nrf2 transcription factor contributes to the induction of alpha-class GST isoenzymes in liver of acute cadmium or manganese intoxicated rats: comparison with the toxic effect on NAD(P)H:quinone reductase. Toxicology 2007; 237:24-34. [PMID: 17573173 DOI: 10.1016/j.tox.2007.04.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 04/19/2007] [Accepted: 04/27/2007] [Indexed: 11/16/2022]
Abstract
In rat liver, in addition to their intrinsic transferase activity, alpha-class GSTs have Se-independent glutathione peroxidase activity toward fatty acid hydroperoxides, cumene hydroperoxide and phospholipids hydroperoxides but not toward H(2)O(2.) We have previously shown that hepatic GST activity by these isoenzymes is significantly increased 24h after cadmium or manganese administration (Casalino et al., 2004). Here it is reported that Se-independent glutathione peroxidase activity by alpha-class GSTs is also stimulated in the liver of intoxicated rats. The stimulation is associated with a higher level of alpha-class GST proteins, whose induction is blocked by actinomycin D co-administration. The observed Se-independent glutathione peroxidase activity is due to alpha-class GST isoenzymes, as indicated by the studies with diethyldithiocarbamate which, at any concentration, equally inhibits both GST and Se-independent glutathione peroxidase and is an uncompetitive inhibitor of both enzymes. As for liver Se-GSPx, it is not at all affected under these toxic conditions. For comparison, we have evaluated the status of another important antioxidant enzyme, NAD(P)H:quinone reductase, 24h after cadmium or manganese administration. NQO1 too results strongly stimulated in the liver of the intoxicated rats. In these animals, a higher expression of Nrf2 protein is observed, actively translocated from the cytoplasm to the nucleus. The results with the transcription inhibitor, actinomycin D, and the effects on Nrf2 protein are the first clear indication that acute manganese intoxication, similarly to that of cadmium and other heavy metals, increases both the hepatic level of Nrf2 and its transfer from the cytoplasm to the nucleus where it actively regulates the induction of phase II enzymes.
Collapse
Affiliation(s)
- Elisabetta Casalino
- Unit of Veterinary Biochemistry, Department of Pharmaco-Biology, University of Bari, Str. Prov. per Casamassima Km 3, 70010 Valenzano, Bari, Italy.
| | - Giovanna Calzaretti
- Unit of Veterinary Biochemistry, Department of Pharmaco-Biology, University of Bari, Str. Prov. per Casamassima Km 3, 70010 Valenzano, Bari, Italy
| | - Matteo Landriscina
- Clinical Oncology Unit, Department of Medical Sciences, University of Foggia, Italy
| | - Cesare Sblano
- Unit of Veterinary Biochemistry, Department of Pharmaco-Biology, University of Bari, Str. Prov. per Casamassima Km 3, 70010 Valenzano, Bari, Italy
| | - Annarita Fabiano
- Clinical Oncology Unit, Department of Medical Sciences, University of Foggia, Italy
| | - Clemente Landriscina
- Unit of Veterinary Biochemistry, Department of Pharmaco-Biology, University of Bari, Str. Prov. per Casamassima Km 3, 70010 Valenzano, Bari, Italy
| |
Collapse
|
16
|
Kim SK, Abdelmegeed MA, Novak RF. Identification of the insulin signaling cascade in the regulation of alpha-class glutathione S-transferase expression in primary cultured rat hepatocytes. J Pharmacol Exp Ther 2005; 316:1255-61. [PMID: 16293713 DOI: 10.1124/jpet.105.096065] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We reported previously that insulin elevated alpha-class glutathione S-transferase (GSTs) protein levels in primary cultured rat hepatocytes (Kim et al., 2003b). In contrast, glucagon down-regulated alpha- and pi-class GST expression, and mechanistic research implicated cAMP and protein kinase A in this process (Kim et al., 2003b). The present study examines the signaling pathways involved in the regulation of alpha-class GST in response to insulin in primary cultured rat hepatocytes. Protein levels of GSTA1/2 and GSTA3/5 and activity of GST toward 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole (NBD) were increased in an insulin concentration-dependent manner. Treatment of cells with the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one] or rapamycin, an inhibitor of mammalian target of rapamycin and ribosomal p70 S6 kinase (p70S6K) phosphorylation, or with an adenovirus containing green fluorescent protein and a dominant-negative and kinase-dead Akt, effectively inhibited the insulin-mediated increase in alpha-class GST expression and GST activity toward NBD. In contrast, PD98059 (2'-amino-3'-methoxyflavone), an inhibitor of mitogen-activated protein kinase kinase, SP600125 (1,9-pyrazoloanthrone), an inhibitor of c-Jun N-terminal kinase, SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imadazole], an inhibitor of p38 mitogen-activated protein kinase, or bisindolylmaleimide, a broad spectrum inhibitor of protein kinase C, did not inhibit the insulin-mediated increase in alpha-class GST protein levels in hepatocytes. These results show that PI3K/Akt/p70S6K signaling is active in the insulin-mediated up-regulation of the antioxidant defense system and that low insulin levels, as encountered in diabetes, potentially increase the susceptibility of hepatocytes to xenobiotic-mediated and/or oxidative stress-mediated damage.
Collapse
Affiliation(s)
- Sang K Kim
- Institute of Environmental Health Sciences, Wayne State University, 2727 South Avenue, Room 4000, Detroit, MI 48201, USA
| | | | | |
Collapse
|
17
|
Tsai CW, Chen HW, Yang JJ, Liu KL, Lii CK. Sulfur amino acid restriction induces the pi class of glutathione S-transferase expression in primary rat hepatocytes. J Nutr 2005; 135:1034-9. [PMID: 15867277 DOI: 10.1093/jn/135.5.1034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The regulation of genes by amino acids is attracting increasing attention. In the present study, we investigated the restriction of expression of the pi class of glutathione S-transferase (GST Yp) by sulfur amino acids. Hepatocytes isolated from male Sprague-Dawley rats were cultured with L-15-based medium containing low (LSAA; 0.1 mmol/L L-methionine and 0.1 mmol/L L-cysteine) or high (HSAA; 0.5 mmol/L L-methionine and 0.2 mmol/L L-cysteine) amounts of sulfur amino acids for up to 6 d. Cellular protein contents did not differ between LSAA- and HSAA-treated cells over the entire period. In contrast, glutathione concentrations were suppressed by the LSAA medium and on d 6 were only 20% of those of HSAA-treated cells (P < 0.05). As shown by immunoblot analysis, GST Yp protein levels were greater in LSAA-treated cells than in HSAA-treated cells (P < 0.05). The induction of GST Yp by L-methionine and L-cysteine restriction was not affected by insulin and dexamethasone, but the latter suppressed GST Yp expression (P < 0.05). LSAA increased GST Yp mRNA levels and GST activity toward ethacrynic acid (P < 0.05). GST Yp induction occurred only in cells with a limited supply of L-methionine; restriction of L-isoleucine, L-leucine, L-lysine, and L-phenylalanine had no significant effect. In contrast with the induction of GST Yp, the expression of the GST isoforms Ya and Yb was not changed by amino acid restriction. In conclusion, hepatic GST Yp gene expression is upregulated by a limited availability of sulfur amino acids.
Collapse
Affiliation(s)
- Chia-Wen Tsai
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
18
|
Ronchi VP, Conde RD, Guillemot JC, Sanllorenti PM. The mouse liver content of carbonic anhydrase III and glutathione S-tranferases A3 and P1 depend on dietary supply of methionine and cysteine. Int J Biochem Cell Biol 2005; 36:1993-2004. [PMID: 15203113 DOI: 10.1016/j.biocel.2004.02.019] [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: 05/20/2003] [Revised: 02/20/2004] [Accepted: 02/25/2004] [Indexed: 11/23/2022]
Abstract
The contents of glutathione S-transferase (GST) subunits, carbonic anhydrase III (CAIII), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and a 230 kDa protein are affected by protein deprivation in mouse liver. In order to know if particular amino acids control these contents, the effects of feeding for 5 days with diets containing different amino acids were examined. After an exploration using SDS-PAGE analysis, the action of selected diets was further examined by distinct techniques. The 230 kDa protein was identified as fatty acid synthase (FAS) by both mass spectrometry and amino acid sequence analyses. Dietary tests showed that: (1) a protein-free diet (PFD) increased the content of glutathione S-transferases P1 and M1, and glyceraldehyde-3-phosphate dehydrogenase, while the content of glutathione S-transferase A3, fatty acid synthase and carbonic anhydrase III decreased; (2) a protein-free diet having either methionine or cysteine preserved the normal contents of glutathione S-transferases P1, A3, M1 and carbonic anydrase III; (3) a protein-free diet having threonine preserved partially the normal contents of glutathione S-transferases P1, A3, M1 and carbonic anhydrase III; (4) a protein-free diet having methionine, threonine and cysteine prevented in part the loss of fatty acid synthase; and (5) the glyceraldehyde-3-phosphate dehydrogenase content was controlled by increased carbohydrate level and/or by lower amino acid content of diets, but not by any specific amino acid. These data indicate that methionine and cysteine exert a main role on the control of liver glutathione S-transferases A3 and P1, and carbonic anhydrase III. Thus, they emerge necessary to prevent unsafe alterations of liver metabolism caused by protein deprivation.
Collapse
Affiliation(s)
- Virginia Paola Ronchi
- Facultad de Ciencias Exactas y Naturales, Instituto de Investigaciones Biológicas, Universidad Nacional de Mar del Plata, C.C. 1245, B7600GTQ Mar del Plata, Argentina.
| | | | | | | |
Collapse
|
19
|
Abstract
This study was designed to determine the oxidative stress induced by nickel sulfate in the liver in the protein-deficient rats. Nickel sulfate in the dose of 800 mg/L in drinking water was administrated to Sprauge Dawley (SD) rats as well as protein-deficient rats for a total duration of 8 weeks. The effects of nickel treatment and protein deficiency separately and in combination were studied on rat liver antioxidant defense system enzymes like catalase, glutathione peroxidase (GPX), glutathione reductase (GR), superoxide dismutase (SOD), reduced glutathione (GSH), and glutathione-S-transferase (GST), as well as on lipid peroxidation (LPO). The investigations revealed a significant increase in the activity of enzymes, which include catalase, Gpx, GR and GST, and in the levels with LPO following nickel treatment in combination with protein deficiency. On the contrary, feeding to control rats resulted in a significant depression in the levels of SOD and GSH. However, nickel treatment to normal rats caused a significant increase in the activity of enzymes catalase and GST and in the levels of LPO, whereas the levels of GSH get significantly depressed. Further, nickel treatment to protein-deficient rats did not cause any additional alteration in the status of liver antioxidants as were observed in conditions of protein deficiency.
Collapse
|
20
|
Kim SG, Kim SO. Pkc downstream of pi3-kinase regulates peroxynitrite formation for nrf2-mediated gsta2 induction. Arch Pharm Res 2004; 27:757-62. [PMID: 15357004 DOI: 10.1007/bf02980145] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The protective adaptive response to electrophiles and reactive oxygen species is mediated by the induction of phase II detoxifying genes including glutathione S-transferases (GSTs). NF-E2-related factor-2 (Nrf2) phosphorylation by protein kinase C (PKC) is a critical event for its nuclear translocation in response to oxidative stress. Previously, we have shown that peroxynitrite plays a role in activation of Nrf2 and Nrf2 binding to the antioxidant response element (ARE) via the pathway of phosphatidylinositol 3-kinase (Pl3-kinase) and that nitric oxide synthase in hepatocytes is required for GSTA2 induction. In view of the importance of PKC and Pl3-kinase in Nrf2-mediated GST induction, we investigated the role of these kinases in peroxynitrite formation for GSTA2 induction by oxidative stress and determined the relationship between PKC and Pl3-kinase. Although PKC activation by phorbol 12-myristate-13-acetate (PMA) did not increase the extents of constitutive and inducible GSTA2 expression, either PKC depletion by PMA or PKC inhibition by staurosporine significantly inhibited GSTA2 induction by tert-butylhydroquinone (t-BHQ) a prooxidant chemical. Therefore, the basal PKC activity is requisite for GSTA2 induction. 3-Morpholinosydnonimine (SIN-1), which decomposes and yields peroxynitrite, induced GSTA2, which was not inhibited by PKC depletion, but slightly enhanced by PKC activation, suggesting that PKC promotes peroxynitrite formation for Nrf2-mediated GSTA2 induction. Treatment of cells with S-nitroso-N-acetyl-penicillamine (SNAP), an exogenous NO donor, in combination with t-BHQ may produce peroxynitrite. GSTA2 induction by SNAP + t-BHQ was not decreased by PKC depletion, but rather enhanced by PKC activation, showing that the activity of PKC might be required for peroxynitrite formation. LY294002 a Pl3-kinase inhibitor blocked GSTA2 induction by t-BHQ, which was reversed by PMA-induced PKC activation. These results provide evidence that PKC may play a role in formation of peroxynitrite that activates Nrf2 for GSTA2 induction and that PKC may serve an activator for GSTA2 induction downstream of Pl3-kinase.
Collapse
Affiliation(s)
- Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea.
| | | |
Collapse
|
21
|
Kim YG, Kim SK, Kwon JW, Park OJ, Kim SG, Kim YC, Lee MG. Effects of cysteine on amino acid concentrations and transsulfuration enzyme activities in rat liver with protein-calorie malnutrition. Life Sci 2003; 72:1171-81. [PMID: 12505547 DOI: 10.1016/s0024-3205(02)02366-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The changes in amino acid concentrations and transsulfuration enzyme activities in liver were investigated after 4-week fed on 23% casein diet (control group) and 5% casein diet without (protein-calorie malnutrition, PCM group) or with (PCMC group) oral administration of cysteine, 250 mg/kg (twice daily, starting from the fourth week) using rats as an animal model. By supplementation with cysteine in PCM rats (PCMC group), cysteine level was elevated almost close to the control level, and glutathione (GSH), aspartic acid and serine levels were restored greater than the control levels. The measurement of transsulfuration enzyme activities exhibited that gamma-glutamylcysteine ligase (gamma-GCL) activity was up-regulated in rats with protein restriction (PCM group), and cysteine supplementation (PCMC group) down-regulated to the control level. One-week supplementation of cysteine (PCMC group) significantly down-regulated the cysteine sulfinate decarboxylase activity. These results indicate that the availability of sulfur amino acid(s) especially cysteine appears to play a role in determining the flux of cysteine between cysteine catabolism and GSH synthesis.
Collapse
Affiliation(s)
- Yoon G Kim
- Department of Pharmacology, College of Medicine, Dankook University, San 29, Anseo-Dong, Chonan, Chungcheongnam-Do 330-714, South Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Azosemide is used in the treatment of oedematous states and hypertension. The exact mechanism of action is not fully understood, but it mainly acts on both the medullary and cortical segments of the thick ascending limb of the loop of Henle. Delayed tolerance was demonstrated in humans by homeostatic mechanisms (principally an increase in aldosterone secretion and perhaps also an increase in the reabsorption of solute in the proximal tubule). After oral administration to healthy humans in the fasting state, the plasma concentration of azosemide reached its peak at 3-4 h with an absorption lag time of approximately 1 h and a terminal half-life of 2-3 h. The estimated extent of absolute oral bioavailability in humans was approximately 20.4%. After oral administration of the same dose of azosemide and furosemide, the diuretic effect was similar between the two drugs, but after intravenous administration, the effect of azosemide was 5.5-8 times greater than that in furosemide. This could be due to the considerable first-pass effect of azosemide. The protein binding to 4% human serum albumin was greater than 95% at azosemide concentrations ranging from 10 to 100 microg/ml using an equilibrium dialysis technique. The poor affinity of human tissues to azosemide was supported by the relatively small value of the apparent post-pseudodistribution volume of distribution (Vdbeta), 0.262 l/kg. Eleven metabolites (including degraded products) of azosemide including M1, glucuronide conjugates of both M1 and azosemide, thiophenemethanol, thiophencarboxylic acid and its glycine conjugate were obtained in rats. Only azosemide and its glucuronide were detected in humans. In humans, total body clearance, renal clearance and terminal half-life of azosemide were 112 ml/min, 41.6 ml/min and 2.03 h, respectively. Azosemide is actively secreted in the renal proximal tubule possibly via nonspecific organic acid secretory pathway in humans. Thus, the amount of azosemide that reaches its site of action could be significantly modified by changes in the capacity of this transport system. This capacity, in turn, could be predictably changed in disease states, resulting in decreased delivery of the diuretic to the transport site, as well as in the presence of other organic acids such as nonsteroidal anti-inflammatory drugs which could compete for active transport of azosemide. The urinary excretion rate of azosemide could be correlated well to its diuretic effects since the receptors are located in the loop of Henle. The diuretic effects of azosemide were dependent on the rate and composition of fluid replacement in rabbits; therefore, this factor should be considered in the evaluation of bioequivalence assessment.
Collapse
Affiliation(s)
- Ok K Suh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | |
Collapse
|
23
|
Kang KW, Choi SH, Kim SG. Peroxynitrite activates NF-E2-related factor 2/antioxidant response element through the pathway of phosphatidylinositol 3-kinase: the role of nitric oxide synthase in rat glutathione S-transferase A2 induction. Nitric Oxide 2002; 7:244-53. [PMID: 12446173 DOI: 10.1016/s1089-8603(02)00117-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The protective adaptive response to electrophiles and reactive oxygen species is mediated by the induction of phase II detoxifying genes through antioxidant response elements (AREs). Our previous study showed that sulfur amino acid deprivation (SAAD) produces peroxides and induces rat glutathione S-transferase A2 (rGSTA2) through NF-E2-related factor 2 (Nrf2)/ARE activation via the pathway of phosphatidylinositol 3-kinase (PI3-kinase). The current study was designed to investigate the role of peroxynitrite in Nrf2/ARE activation and rGSTA2 induction. L-Arginine deficiency or N(G)-nitro-L-arginine methyl ester (L-NAME) reduced peroxide production induced by SAAD in H4IIE cells. Northern and Western blot analyses revealed that the levels of rGSTA2 mRNA and protein were significantly increased 24h after incubation of the cells in SAAD medium, which was inhibited by L-arginine deficiency or L-NAME. Subcellular fractionation and gel shift analyses revealed that SAAD increased the level of nuclear Nrf2 and activated ARE, which were also blocked by L-arginine deficiency or L-NAME. Whereas the exogenous NO donor S-nitroso-N-acetyl-penicillamine (SNAP) alone failed to significantly induce rGSTA2, SNAP enhanced SAAD-inducible rGSTA2 expression, verifying the notion that peroxynitrite derived from NO contributes to rGSTA2 induction. 3-Morpholinosydnonimine (SIN-1), which decomposes and yields peroxynitrite, increased the rGSTA2 mRNA and protein levels in a dose-dependent manner. SIN-1 increased the level of nuclear Nrf2 and activated Nrf2/ARE, which was supershifted by anti-Nrf2 and anti-Maf antibodies. SIN-1 increased the activity of PI3-kinase, as monitored by phosphorylation of Akt. SIN-1-inducible rGSTA2 expression was inhibited by PI3-kinase inhibitors. These results provide evidence that peroxynitrite plays an essential role in nuclear translocation of Nrf2 and ARE activation through the pathway of PI3-kinase and that nitric oxide synthase is involved in the induction of rGSTA2.
Collapse
Affiliation(s)
- Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Sillim-dong, Kwanak-gu, 151-742, Seoul, Republic of Korea
| | | | | |
Collapse
|
24
|
Kang KW, Novak RF, Lee CH, Kim SG. Induction of microsomal epoxide hydrolase by sulfur amino acid deprivation via the pathway of C-Jun N-terminal kinase and its extracellular exposure during cell death. Free Radic Biol Med 2002; 32:1017-32. [PMID: 12008117 DOI: 10.1016/s0891-5849(02)00788-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Microsomal epoxide hydrolase (mEH), an epoxide detoxifying enzyme and putative cell surface autoantigen, is inducible by xenobiotics and by certain pathophysiological conditions (e.g., tumorigenesis and protein-calorie malnutrition). The present study was designed to determine mEH expression in H4IIE cells during cell death initiated by sulfur amino acid deprivation (SAAD) and to identify the signaling pathway for the enzyme induction. SAAD induced cell death at 48-72 h with translocation of Bax to mitochondria and increased mitochondrial permeability with cytochrome c release, both of which were prevented by SB203580 or by dominant-negative JNK1 [JNK1(-)] stable transfection. Caspase-3 activity was only marginally increased by SAAD. Neither genomic DNA fragmentation nor poly(ADP-ribose) polymerase cleavage was observed during SAAD-induced cell death. Thus, SAAD induced cell death independent of caspase activation. This was supported by the observation that benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, a general caspase inhibitor, did not prevent cell death. The levels of mEH mRNA and protein were notably increased in cells under SAAD for 48-72 h. The induction of mEH occurred in parallel with cell death. Whereas SAAD-induced cell death resulted from both JNK1 and p38 kinase activation, mEH induction was decreased only by JNK1(-) transfection. Immunocytochemistry revealed that mEH protein was intensely stained in dying cells, cellular fragments and cell debris. Furthermore, the number of cells positive for surface mEH substantially increased by SAAD, as evidenced by flow cytometry analysis. These results demonstrated that SAAD induced nonapoptotic cell death with Bax translocation to mitochondria and mitochondrial cytochrome c release, but not through caspase-3 activation, and that mEH was induced by SAAD via the pathway of JNK1, but not ERK1/2 or p38 kinase, in parallel with cell death.
Collapse
Affiliation(s)
- Keon Wook Kang
- National Research Laboratory (MDT), College of Pharmacy, Seoul National University, Sillim-dong Kwanak-gu, Seoul 151-742, South Korea
| | | | | | | |
Collapse
|
25
|
Kim HJ, Kim SG. Alterations in cellular Ca(2+) and free iron pool by sulfur amino acid deprivation: the role of ferritin light chain down-regulation in prooxidant production. Biochem Pharmacol 2002; 63:647-57. [PMID: 11992632 DOI: 10.1016/s0006-2952(01)00877-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Deficiency of sulfur amino acids occurs in certain pathophysiological states such as protein-calorie malnutrition. Sulfur amino acid deprivation (SAAD) increases oxidative stress through a decrease in GSH. Ferritin expression is induced by oxidative stress, which confers resistance to oxidative insults. The effects of SAAD on the changes in cellular Ca(2+) and free iron pool, prooxidant production and the ferritin light chain (FLC) expression were comparatively evaluated in Hepa1c1c7 and Raw264.7 cells. [Ca(2+)](i) was rapidly increased by SAAD. Sulfhydryl-containing compounds prevented the increase in [Ca(2+)](i) in cells under SAAD, supporting the role of redox-state in the regulation of [Ca(2+)](i). Thapsigargin or Ca(2+)-free medium inhibited the increase in [Ca(2+)](i), showing that Ca(2+) originated from endoplasmic reticulum as well as from extracellular source. Inhibition of Ca(2+) mobilization decreased the fluorescence of Phen Green SK inside cells, representing the inhibition of free iron release. Both inhibition of Ca(2+) mobilization and iron chelation decreased dichlorofluorescein oxidation, indicating the possibility that the increase in [Ca(2+)](i) affected that in cellular free iron and prooxidant production. FLC protein level was immunochemically detectable in Raw264.7 cells, but not in Hepa1c1c7 cells. SAAD alone (or in combination with FeSO(4)) down-regulated FLC protein expression, while SAAD increased the FLC mRNA level in both Hepa1c1c7 and Raw264.7 cells. Calcium or iron chelators prevented increases in the FLC mRNA. These results provided evidence that changes in cellular Ca(2+) and iron pool by SAAD increased cellular oxidative stress and that the down-regulation of FLC protein by SAAD would further enhance prooxidant production in spite of the increase in FLC mRNA.
Collapse
Affiliation(s)
- Hye Jung Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 151-742, Seoul, South Korea
| | | |
Collapse
|
26
|
Lee AK, Kang KW, Kim YG, Cho MK, Lee MG, Shim CK, Chung SJ, Kim SG. Identification of genes enhanced by protein-calorie malnutrition by differential display polymerase chain reaction (expression of fibrinogen B beta chain, B cell translocation gene 1 and thyroid hormone responsive protein genes). Mol Cell Biochem 2002; 231:163-71. [PMID: 11952159 DOI: 10.1023/a:1014481319278] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Protein-calorie malnutrition (PCM), as one of global health problems, arises during protein and/or energy deficit due to disease and nutritional inadequacy. Previously, we showed that PCM elicited oxidative stress with activation of the phase II detoxifying gene expression, which was reversed by cysteine supplementation. As part of the attempts to identify the cellular adaptive responses and the associated gene expression during PCM, the current study was initiated to analyze the genes differentially expressed in the rat during PCM. Among 1,916 bands amplified, 85 putative differentially amplified bands were enhanced by PCM in the liver, while the expression of 64 bands was suppressed. Northern and/or reverse transcription-polymerase chain reaction (RT-PCR) analyses revealed that PCM increased the expression of fibrinogen B beta chain, B cell translocation gene I (BTGI) and thyroid hormone responsive protein (THRP) mRNAs. The increase in the hepatic fibrinogen B beta chain mRNA was not prevented by cysteine supplementation, whereas cysteine decreased the enhancement in the rGSTA2 and microsomal epoxide hydrolase mRNA expression. Cysteine was also active in reversing the increase in BTG1 mRNA during PCM. This was supported by the increase in BTG1 mRNA in H4IIE cells exposed to sulfur amino acid-deprived medium. Northern blot analysis revealed that THRP, highly expressed in the brain in a tissue-specific manner, was induced by PCM and that cysteine supplementation abolished the THRP induction. Conversely, the level of hepatic albumin mRNA was markedly decreased by PCM, which was partially restored by cysteine supplementation. Differential display RT-PCR analysis allowed us to identify the genes that are responsive to oxidative stress during PCM and to characterize the differential role of cysteine on the expression of the fibrinogen B beta chain, BTG1 and THRP genes as a homeostatic adaptive response during protein deficiency.
Collapse
Affiliation(s)
- Ae Kyung Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Son MH, Kang KW, Lee CH, Kim SG. Potentiation of cadmium-induced cytotoxicity by sulfur amino acid deprivation through activation of extracellular signal-regulated kinase1/2 (ERK1/2) in conjunction with p38 kinase or c-jun N-terminal kinase (JNK). Complete inhibition of the potentiated toxicity by U0126 an ERK1/2 and p38 kinase inhibitor. Biochem Pharmacol 2001; 62:1379-90. [PMID: 11709198 DOI: 10.1016/s0006-2952(01)00780-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The mechanisms of cadmium-induced toxicity may include oxidative stress, altered redox homeostasis, and injuries to organelles. The current study was designed to study the effect of decreased cellular glutathione (GSH) content by sulfur amino acid deprivation on cadmium toxicity and to identify the signaling pathways responsible for the cytotoxicity. GSH content was increased by cadmium in H4IIE cells prior to cell death, which was prevented by excess GSH or cysteine. Cell viability, however, was not improved by GSH or cysteine complexation of cadmium. Cadmium-induced cytotoxicity was 40-fold potentiated in cells with decreased GSH by sulfur amino acid deprivation. Cadmium in combination with decreased GSH markedly increased apoptotic cell death. Mitogen-activated protein kinases including extracellular signal-regulated kinase 1/2, p38 kinase and c-Jun N-terminal kinase (JNK) were all activated 1-12 hr after sulfur amino acid deprivation. U0126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene), which inhibited activation of extracellular signal-regulated kinase1/2 and p38 kinase in cells under sulfur amino acid deprivation, completely prevented potentiation in Cd-induced cytotoxicity and apoptosis. Potentiation of cadmium toxicity by sulfur amino acid deprivation was prevented in part by either PD98059 or SB203580, or in cells stably expressing dominant negative mutant of JNK1, and to greater extents by PD98059 in combination with either SB203580 or JNK1(-) transfection. These results demonstrated that decreased cellular GSH content potentiated cytotoxicity induced by cadmium at the level of human exposure, and that the potentiation of cytotoxicity resulted from activation of extracellular signal-regulated kinase1/2 in conjunction with p38 kinase or JNK.
Collapse
Affiliation(s)
- M H Son
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Sillim-dong, Kwanak-gu, 151-742, Seoul, South Korea
| | | | | | | |
Collapse
|
28
|
Son MH, Kang KW, Lee CH, Kim SG. Potentiation of arsenic-induced cytotoxicity by sulfur amino acid deprivation (SAAD) through activation of ERK1/2, p38 kinase and JNK1: the distinct role of JNK1 in SAAD-potentiated mercury toxicity. Toxicol Lett 2001; 121:45-55. [PMID: 11312036 DOI: 10.1016/s0378-4274(01)00314-9] [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: 10/18/2022]
Abstract
Sulfur amino acid deficiency occurs in certain pathophysiological situations (e.g. protein-calorie malnutrition). Previous studies revealed that sulfur amino acid deprivation (SAAD) activated MAP kinases and potentiated cadmium-induced cytotoxicity by activation of ERK1/2 in conjunction with p38 kinase or JNK. The present study was designed to determine susceptibility of cells to a variety of heavy metals in combination with SAAD. Viability was assessed in H4IIE cells treated with sodium arsenite, mercuric chloride, sodium selenite, lead acetate, chromium trioxide or manganese chloride. SAAD potentiated the cytotoxicity of H4IIE cells by arsenic or mercury (i.e. EC50, 19 and 5 microM in SAAD vs. 401 and 42 microM in control medium, respectively). TUNEL assays revealed that the potentiated arsenic or mercury toxicity involved apoptotic cell death. Lead or selenite moderately elicited cell death, which was not enhanced by SAAD. Chromium or manganese caused no significant cytotoxicity. Treatment of cells with U0126 [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene] an ERK1/2 inhibitor or SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole] a p38 kinase inhibitor effectively prevented SAAD-potentiated arsenic toxicity. The potentiated arsenic toxicity was also inhibited in cells stably expressing a dominant negative mutant of c-Jun N-terminal kinase 1 [JNK1(-)]. The inhibitors of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 kinase failed to prevent mercury-induced toxicity enhanced by SAAD. JNK1(-) cells were minimally susceptible to mercury in SAAD medium. These results demonstrated that SAAD potentiated cytotoxicity induced by arsenic or mercury and that activation of ERK1/2, p38 kinase and JNK1 was responsible for the potentiated arsenic toxicity, whereas the mercury toxicity enhanced by SAAD was mediated with the activity of JNK1.
Collapse
Affiliation(s)
- M H Son
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Sillim-dong, Kwanak-gu, 151-742, Seoul, South Korea
| | | | | | | |
Collapse
|
29
|
Kang KW, Ryu JH, Kim SG. The essential role of phosphatidylinositol 3-kinase and of p38 mitogen-activated protein kinase activation in the antioxidant response element-mediated rGSTA2 induction by decreased glutathione in H4IIE hepatoma cells. Mol Pharmacol 2000; 58:1017-25. [PMID: 11040049 DOI: 10.1124/mol.58.5.1017] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The protective adaptive response to electrophiles and reactive oxygen species is mediated by the enhanced expression of the phase II detoxifying genes through antioxidant response elements (AREs). The current study was designed to identify the signaling pathways responsible for the expression of rGSTA2 in response to cellular oxidative stress and to establish the molecular mechanistic basis. Deprivation of cystine and methionine caused oxidative stress in H4IIE hepatoma cells as evidenced by a marked decrease in the reduced glutathione (first order rate constant = 0.056 h(-1); t(1/2) = 12.6 h) and an increase in pro-oxidant production. Electrophoretic mobility shift assay revealed that the ARE complex, consisting of Nrf-1/2 and Maf proteins, was activated 12 to 48 h after sulfur amino acid deprivation (SAAD). The rGSTA2 mRNA level was elevated by SAAD beginning at 24 h, whereas the rGSTA2 subunit was maximally induced at 48 h. Nuclear ARE activation and rGSTA2 mRNA increase were both completely inhibited by wortmannin or LY294002, the phosphatidylinositol 3-kinase (PI3-kinase) inhibitors. The p38 mitogen-activated protein (MAP) kinase was activated at 0.5 to 3 h after SAAD, followed by sustained diminished activation up to 12 h. Inhibition of p38 MAP kinase by SB203580 prevented the ARE-mediated rGSTA2 induction. The activation of p38 MAP kinase, however, failed to be inhibited by wortmannin or LY294002, showing that PI3-kinase is not involved in the activation of p38 MAP kinase. Data showed that PI3-kinase plays an essential role in the ARE-mediated rGSTA2 induction by oxidative stress after SAAD, which activates the p38 MAP kinase and leads to rGSTA2 induction.
Collapse
Affiliation(s)
- K W Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | | | | |
Collapse
|