1
|
Haider M, Sharma S, Agrahari AK, Dikshit M, Pathak DP, Asthana S. Crystallographic mining driven computer-guided approach to identify the ASK1 inhibitor likely to perturb the catalytic region. J Biomol Struct Dyn 2025; 43:1290-1304. [PMID: 38069610 DOI: 10.1080/07391102.2023.2291545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/18/2023] [Indexed: 01/16/2025]
Abstract
The pathological levels of reactive oxygen species (ROS) and oxidative stress has been recognized as a critical driver for inflammatory disorders. Apoptosis signal-regulating kinase 1 (ASK1) has been reported to be activated by intracellular ROS and its inhibition leads to a down regulation of p38-and JNK-dependent signaling. ASK1 inhibitors are reported to have the potential to treat clinically important inflammatory pathologies including liver, pulmonary and renal disorders. In view of its biological and pathological significance, inhibition of ASK1 with small molecules has been pursued as an attractive strategy to combat human diseases such as non-alcoholic steatohepatitis (NASH). Despite several ASK1 inhibitors being developed, the failure in Phase 3 clinical trials of most advanced candidate selonsertib's, underscores to discover therapeutic agents with diverse chemical moiety. Here, by using structural pharmacophore and enumeration strategy on mining co-crystals of ASK1, different scaffolds were generated to enhance the chemical diversity keeping the critical molecular interaction in the catalytic site intact. A total of 15,772 compounds were generated from diverse chemical scaffolds and were evaluated using a virtual screening pipeline. Based on docking and MM-GBSA scores, a lead candidate, S3C-1-D424 was identified from top hits. A comparative molecular dynamics simulations (MD) of APO, Selonsertib and shortlisted potential candidates combined with pharmacokinetics profiling and thermodynamic analysis, demonstrating their suitability as potential ASK1 inhibitors to explore further for establishment towards hit-to-lead campaign.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohamad Haider
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSR University, New Delhi, India
| | - Shilpa Sharma
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Ashish Kumar Agrahari
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Madhu Dikshit
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Pharmacology Division, Central Drug Research Institute, Lucknow, India
| | - Dharam Pal Pathak
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSR University, New Delhi, India
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| |
Collapse
|
2
|
Park SY, Kim KS, Lee WY, Kim CE, Lee S. Integrative Approach to Identifying System-Level Mechanisms of Chung-Sang-Bo-Ha-Hwan's Influence on Respiratory Tract Diseases: A Network Pharmacological Analysis with Experimental Validation. PLANTS (BASEL, SWITZERLAND) 2023; 12:3024. [PMID: 37687271 PMCID: PMC10489874 DOI: 10.3390/plants12173024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
Chung-Sang-Bo-Ha-Hwan (CSBHH) is an herbal prescription widely used to treat various chronic respiratory diseases. To investigate the system-level treatment mechanisms of CSBHH in respiratory tract diseases, we identified 56 active ingredients of CSBHH and evaluated the degree of overlap between their targets and respiratory tract disease-associated proteins. We then investigated the respiratory tract disease-related signaling pathways associated with CSBHH targets. Enrichment analysis showed that the CSBHH targets were significantly associated with various signaling pathways related to inflammation, alveolar structure, and tissue fibrosis. Experimental validation was conducted using phorbol-12-myristate-13-acetate (PMA)-stimulated NCI-H292 cells by analyzing the mRNA expression levels of biomarkers (IL-1β and TNF-α for inflammation; GSTP1, GSTM1, and PTEN for apoptosis) derived from network pharmacological analysis, in addition to the mucin genes MUC5AC and MUC2, to investigate the phlegm-expelling effect of CSBHH. The mRNA expression levels of these genes were consistent with network pharmacological predictions in a concentration-dependent manner. These results suggest that the therapeutic mechanisms of CSBHH in respiratory tract diseases could be attributed to the simultaneous action of multiple active ingredients in the herbal prescription.
Collapse
Affiliation(s)
- Sa-Yoon Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Kang-Sub Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea;
| | - Won-Yung Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
3
|
Zhang C, Lu LF, Li ZC, Han KJ, Wang XL, Chen DD, Xiong F, Li XY, Zhou L, Ge F, Li S. Zebrafish MAP2K7 Simultaneously Enhances Host IRF7 Stability and Degrades Spring Viremia of Carp Virus P Protein via Ubiquitination Pathway. J Virol 2023; 97:e0053223. [PMID: 37367226 PMCID: PMC10373533 DOI: 10.1128/jvi.00532-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023] Open
Abstract
During viral infection, host defensive proteins either enhance the host immune response or antagonize viral components directly. In this study, we report on the following two mechanisms employed by zebrafish mitogen-activated protein kinase kinase 7 (MAP2K7) to protect the host during spring viremia of carp virus (SVCV) infection: stabilization of host IRF7 and degradation of SVCV P protein. In vivo, map2k7+/- (map2k7-/- is a lethal mutation) zebrafish showed a higher lethality, more pronounced tissue damage, and more viral proteins in major immune organs than the controls. At the cellular level, overexpression of map2k7 significantly enhanced host cell antiviral capacity, and viral replication and proliferation were significantly suppressed. Additionally, MAP2K7 interacted with the C terminus of IRF7 and stabilized IRF7 by increasing K63-linked polyubiquitination. On the other hand, during MAP2K7 overexpression, SVCV P proteins were significantly decreased. Further analysis demonstrated that SVCV P protein was degraded by the ubiquitin-proteasome pathway, as the attenuation of K63-linked polyubiquitination was mediated by MAP2K7. Furthermore, the deubiquitinase USP7 was indispensable in P protein degradation. These results confirm the dual functions of MAP2K7 during viral infection. IMPORTANCE Normally, during viral infection, host antiviral factors individually modulate the host immune response or antagonize viral components to defense infection. In the present study, we report that zebrafish MAP2K7 plays a crucial positive role in the host antiviral process. According to the weaker antiviral capacity of map2k7+/- zebrafish than that of the control, we find that MAP2K7 reduces host lethality through two pathways, as follows: enhancing K63-linked polyubiquitination to promote host IRF7 stability and attenuating K63-mediated polyubiquitination to degrade the SVCV P protein. These two mechanisms of MAP2K7 reveal a special antiviral response in lower vertebrates.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ke-Jia Han
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xue-Li Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Feng Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xi-Yin Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Feng Ge
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
4
|
Li Y, Liu T, Li X, Yang M, Liu T, Bao J, Jiang M, Hu L, Wang Y, Shao P, Jiang J. Combined surface functionalization of MSC membrane and PDA inhibits neurotoxicity induced by Fe 3O 4 in mice based on apoptosis and autophagy through the ASK1/JNK signaling pathway. Aging (Albany NY) 2023; 15:6933-6949. [PMID: 37470690 PMCID: PMC10415563 DOI: 10.18632/aging.204884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/23/2023] [Indexed: 07/21/2023]
Abstract
The extensive utilization of iron oxide nanoparticles in medical and life science domains has led to a substantial rise in both occupational and public exposure to these particles. The potential toxicity of nanoparticles to living organisms, their impact on the environment, and the associated risks to human health have garnered significant attention and come to be a prominent area in contemporary research. The comprehension of the potential toxicity of nanoparticles has emerged as a crucial concern to safeguard human health and facilitate the secure advancement of nanotechnology. As nanocarriers and targeting agents, the biocompatibility of them determines the use scope and application prospects, meanwhile surface modification becomes an important measure to improve the biocompatibility. Three different types of iron oxide nanoparticles (Fe3O4, Fe3O4@PDA and MSCM-Fe3O4@PDA) were injected into mice through the tail veins. The acute neurotoxicity of them in mice was evaluated by measuring the levels of autophagy and apoptosis in the brain tissues. Our data revealed that iron oxide nanoparticles could cause nervous system damage by regulating the ASK1/JNK signaling pathway. Apoptosis and autophagy may play potential roles in this process. Exposure to combined surface functionalization of mesenchymal stem cell membrane and polydopamine showed the neuroprotective effect and may alleviate brain nervous system disorders.
Collapse
Affiliation(s)
- Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Yibin Jilin University Research Institute, Jilin University, Yibin, Sichuan, China
| | - Xiuying Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Modi Yang
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Tianxin Liu
- Jilin University School of Public Health, Changchun, Jilin, China
| | - Jindian Bao
- Jilin University School of Public Health, Changchun, Jilin, China
| | - Miao Jiang
- Jilin University School of Public Health, Changchun, Jilin, China
| | - Lingling Hu
- Jilin University School of Public Health, Changchun, Jilin, China
| | - Yuzhuo Wang
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Pu Shao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Choi YJ, Shin MJ, Youn GS, Park JH, Yeo HJ, Yeo EJ, Kwon HJ, Lee LR, Kim NY, Kwon SY, Jung HY, Cho YJ, Kim DW, Park J, Han KH, Lee KW, Park JK, Lee CH, Eum WS, Choi SY. Protective Effects of PEP-1-GSTA2 Protein in Hippocampal Neuronal Cell Damage Induced by Oxidative Stress. Int J Mol Sci 2023; 24:ijms24032767. [PMID: 36769090 PMCID: PMC9917430 DOI: 10.3390/ijms24032767] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
Glutathione S-transferase alpha 2 (GSTA2), a member of the glutathione S-transferase family, plays the role of cellular detoxification against oxidative stress. Although oxidative stress is related to ischemic injury, the role of GSTA2 against ischemia has not been elucidated. Thus, we studied whether GSTA2 prevents ischemic injury by using the PEP-1-GSTA2 protein which has a cell-permeable protein transduction domain. We revealed that cell-permeable PEP-1-GSTA2 transduced into HT-22 cells and markedly protected cell death via the inhibition of reactive oxygen species (ROS) production and DNA damage induced by oxidative stress. Additionally, transduced PEP-1-GSTA2 promoted mitogen-activated protein kinase (MAPK), and nuclear factor-kappaB (NF-κB) activation. Furthermore, PEP-1-GSTA2 regulated Bcl-2, Bax, cleaved Caspase-3 and -9 expression protein levels. An in vivo ischemic animal model, PEP-1-GSTA2, markedly prevented the loss of hippocampal neurons and reduced the activation of microglia and astrocytes. These findings indicate that PEP-1-GSTA2 suppresses hippocampal cell death by regulating the MAPK and apoptotic signaling pathways. Therefore, we suggest that PEP-1-GSTA2 will help to develop the therapies for oxidative-stress-induced ischemic injury.
Collapse
Affiliation(s)
- Yeon Joo Choi
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Gi Soo Youn
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jung Hwan Park
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Eun Ji Yeo
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyun Jung Kwon
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Lee Re Lee
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Na Yeon Kim
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Su Yeon Kwon
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyo Young Jung
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon 24253, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyu Hyung Han
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Keun Wook Lee
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jong Kook Park
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Chan Hee Lee
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
- Correspondence: (W.S.E.); (S.Y.C.); Tel.: +82-33-248-2112 (W.S.E. & S.Y.C.); Fax: +82-33-248-3202 (W.S.E. & S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
- Correspondence: (W.S.E.); (S.Y.C.); Tel.: +82-33-248-2112 (W.S.E. & S.Y.C.); Fax: +82-33-248-3202 (W.S.E. & S.Y.C.)
| |
Collapse
|
6
|
Dubois-Pot-Schneider H, Aninat C, Kattler K, Fekir K, Jarnouen K, Cerec V, Glaise D, Salhab A, Gasparoni G, Takashi K, Ishida S, Walter J, Corlu A. Transcriptional and Epigenetic Consequences of DMSO Treatment on HepaRG Cells. Cells 2022; 11:cells11152298. [PMID: 35892596 PMCID: PMC9331440 DOI: 10.3390/cells11152298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Dimethyl sulfoxide (DMSO) is used to sustain or favor hepatocyte differentiation in vitro. Thus, DMSO is used in the differentiation protocol of the HepaRG cells that present the closest drug-metabolizing enzyme activities to primary human hepatocytes in culture. The aim of our study is to clarify its influence on liver-specific gene expression. For that purpose, we performed a large-scale analysis (gene expression and histone modification) to determine the global role of DMSO exposure during the differentiation process of the HepaRG cells. The addition of DMSO drives the upregulation of genes mainly regulated by PXR and PPARα whereas genes not affected by this addition are regulated by HNF1α, HNF4α, and PPARα. DMSO-differentiated-HepaRG cells show a differential expression for genes regulated by histone acetylation, while differentiated-HepaRG cells without DMSO show gene signatures associated with histone deacetylases. In addition, we observed an interplay between cytoskeleton organization and EMC remodeling with hepatocyte maturation.
Collapse
Affiliation(s)
- Hélène Dubois-Pot-Schneider
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
- Correspondence: ; Tel.: +33-372746115
| | - Caroline Aninat
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Kathrin Kattler
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Karim Fekir
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Kathleen Jarnouen
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Virginie Cerec
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Denise Glaise
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Abdulrahman Salhab
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Gilles Gasparoni
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Kubo Takashi
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Kawasaki 2109501, Japan; (K.T.); (S.I.)
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Kawasaki 2109501, Japan; (K.T.); (S.I.)
| | - Jörn Walter
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Anne Corlu
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| |
Collapse
|
7
|
Yılmaz C, Othman Pirdawid A, Fidan Babat C, Konuş M, Çetin D, Kıvrak A, Algso MAS, Arslan Ş, Mutlu D, Otur Ç, Kurt Kızıldoğan A. A Thiophene Derivative, 2‐Bromo‐5‐(2‐(methylthio)phenyl)thiophene, Has Effective Anticancer Potential with Other Biological Properties. ChemistrySelect 2022. [DOI: 10.1002/slct.202200784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Can Yılmaz
- Department of Molecular Biology and Genetics Faculty of Science Van Yuzuncu Yil University 65080 Van Turkey
| | - Ahmed Othman Pirdawid
- Department of Molecular Biology and Genetics Faculty of Science Van Yuzuncu Yil University 65080 Van Turkey
| | - Ceylan Fidan Babat
- Department of Molecular Biology and Genetics Faculty of Science Van Yuzuncu Yil University 65080 Van Turkey
| | - Metin Konuş
- Department of Molecular Biology and Genetics Facuty of Arts and Science Hitit University 19030 Çorum Turkey
| | - Doğan Çetin
- Department of Molecular Biology and Genetics Faculty of Science Van Yuzuncu Yil University 65080 Van Turkey
| | - Arif Kıvrak
- Department of Chemistry Facuty of Arts and Sciences Osmangazi University 26040 Eskişehir Turkey
| | - Muheb A. S. Algso
- Department of Chemistry Faculty of Science Van Yuzuncu Yil University 65080 Van Turkey
| | - Şevki Arslan
- Department of Biology Faculty of Arts and Science Pamukkale University 20160 Denizli Turkey
| | - Doğukan Mutlu
- Department of Biology Faculty of Arts and Science Pamukkale University 20160 Denizli Turkey
| | - Çiğdem Otur
- Department of Agricultural Biotechnology Faculty of Agriculture Ondokuz Mayıs University 55270 Samsun Turkey
| | - Aslıhan Kurt Kızıldoğan
- Department of Agricultural Biotechnology Faculty of Agriculture Ondokuz Mayıs University 55270 Samsun Turkey
| |
Collapse
|
8
|
Rojas Á, Lara-Romero C, Muñoz-Hernández R, Gato S, Ampuero J, Romero-Gómez M. Emerging pharmacological treatment options for MAFLD. Ther Adv Endocrinol Metab 2022; 13:20420188221142452. [PMID: 36533188 PMCID: PMC9747889 DOI: 10.1177/20420188221142452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/13/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) prevalence and incidence is rising globally. It is associated with metabolic comorbidities, obesity, overweight, type 2 diabetes mellitus, and at least two metabolic risk factors, such as hypertension, hypertriglyceridemia, hypercholesterolemia, insulin resistance, and cardiovascular risk, increasing the risk of mortality. The excessive accumulation of fat comprises apoptosis, necrosis, inflammation and ballooning degeneration progressing to fibrosis, cirrhosis, and liver decompensations including hepatocellular carcinoma development. The limitation of approved drugs to prevent MAFLD progression is a paradigm. This review focuses on recent pathways and targets with evidence results in phase II/III clinical trials.
Collapse
Affiliation(s)
- Ángela Rojas
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Antonio Maura Montaner s/n, 41013 Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta 0 28029 Madrid, Madrid, Spain
| | - Carmen Lara-Romero
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Rocío Muñoz-Hernández
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Sheila Gato
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Javier Ampuero
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | | |
Collapse
|
9
|
Obsilova V, Honzejkova K, Obsil T. Structural Insights Support Targeting ASK1 Kinase for Therapeutic Interventions. Int J Mol Sci 2021; 22:ijms222413395. [PMID: 34948191 PMCID: PMC8705584 DOI: 10.3390/ijms222413395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/22/2022] Open
Abstract
Apoptosis signal-regulating kinase (ASK) 1, a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family, modulates diverse responses to oxidative and endoplasmic reticulum (ER) stress and calcium influx. As a crucial cellular stress sensor, ASK1 activates c-Jun N-terminal kinases (JNKs) and p38 MAPKs. Their excessive and sustained activation leads to cell death, inflammation and fibrosis in various tissues and is implicated in the development of many neurological disorders, such as Alzheimer’s, Parkinson’s and Huntington disease and amyotrophic lateral sclerosis, in addition to cardiovascular diseases, diabetes and cancer. However, currently available inhibitors of JNK and p38 kinases either lack efficacy or have undesirable side effects. Therefore, targeted inhibition of their upstream activator, ASK1, stands out as a promising therapeutic strategy for treating such severe pathological conditions. This review summarizes recent structural findings on ASK1 regulation and its role in various diseases, highlighting prospects for ASK1 inhibition in the treatment of these pathologies.
Collapse
Affiliation(s)
- Veronika Obsilova
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
- Correspondence: (V.O.); (T.O.); Tel.: +420-325-87-3513 (V.O.); +420-22-195-1303 (T.O.)
| | - Karolina Honzejkova
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 12843 Prague, Czech Republic;
| | - Tomas Obsil
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 12843 Prague, Czech Republic;
- Correspondence: (V.O.); (T.O.); Tel.: +420-325-87-3513 (V.O.); +420-22-195-1303 (T.O.)
| |
Collapse
|
10
|
Geng Y, Wang Y, Sun R, Kang X, Zhao H, Zhu M, Sun Y, Hu Y, Wang Z, Tian X, Zhao Y, Yao J. Carnosol alleviates nonalcoholic fatty liver disease by inhibiting mitochondrial dysfunction and apoptosis through targeting of PRDX3. Toxicol Appl Pharmacol 2021; 432:115758. [PMID: 34678374 DOI: 10.1016/j.taap.2021.115758] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/15/2021] [Accepted: 10/16/2021] [Indexed: 12/19/2022]
Abstract
Mitochondrial dysfunction is a major factor in nonalcoholic fatty liver disease (NAFLD), preceding insulin resistance and hepatic steatosis. Carnosol (CAR) is a kind of diterpenoid with antioxidant, anti-inflammatory and antitumor activities. Peroxiredoxin 3 (PRDX3), a mitochondrial H2O2-eliminating enzyme, undergoes overoxidation and subsequent inactivation under oxidative stress. The purpose of this study was to investigate the protective effect of the natural phenolic compound CAR on NAFLD via PRDX3. Mice fed a high-fat diet (HFD) and AML-12 cells treated with palmitic acid (PA) were used to detect the molecular mechanism of CAR in NAFLD. We found that pharmacological treatment with CAR notably moderated HFD- and PA- induced steatosis and liver injury, as shown by biochemical assays, Oil Red O and Nile Red staining. Further mechanistic investigations revealed that CAR exerted anti-NAFLD effects by inhibiting mitochondrial oxidative stress, perturbation of mitochondrial dynamics, and apoptosis in vivo and in vitro. The decreased protein and mRNA levels of PRDX3 were accompanied by intense oxidative stress after PA intervention. Interestingly, CAR specifically bound PRDX3, as shown by molecular docking assays, and increased the expression of PRDX3. However, the hepatoprotection of CAR in NAFLD was largely abolished by specific PRDX3 siRNA, which increased mitochondrial dysfunction and exacerbated apoptosis in vitro. In conclusion, CAR suppressed lipid accumulation, mitochondrial dysfunction and hepatocyte apoptosis by activating PRDX3, mitigating the progression of NAFLD, and thus, CAR may represent a promising candidate for clinical treatment of steatosis.
Collapse
Affiliation(s)
- Yunfei Geng
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Ruimin Sun
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Xiaohui Kang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Huanyu Zhao
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Meiyang Zhu
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Yu Sun
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Yan Hu
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China.
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
11
|
Ware BR, Liu JS, Monckton CP, Ballinger KR, Khetani SR. Micropatterned Coculture With 3T3-J2 Fibroblasts Enhances Hepatic Functions and Drug Screening Utility of HepaRG Cells. Toxicol Sci 2021; 181:90-104. [PMID: 33590212 DOI: 10.1093/toxsci/kfab018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human liver models are useful for assessing compound metabolism/toxicity; however, primary human hepatocyte (PHH) lots are limited and highly variable in quality/viability. In contrast, cell lines, such as HepaRG, are cheaper and more reproducible surrogates for initial compound screening; however, hepatic functions and sensitivity for drug outcomes need improvement. Here, we show that HepaRGs cocultured with murine embryonic 3T3-J2 fibroblasts, previously shown to induce PHH functions, could address such limitations. We either micropatterned HepaRGs or seeded them "randomly" onto collagen-coated plates before 3T3-J2 coculture. Micropatterned cocultures (HepaRG-MPCCs) secreted 2- to 4-fold more albumin and displayed more stable cytochrome P450 activities than HepaRG conventional confluent monocultures (HepaRG-CCs) and HepaRG micropatterned hepatocytes (HepaRG-MPHs) for 4 weeks, even when excluding dimethyl sulfoxide from the medium. Furthermore, HepaRG-MPCCs had the most albumin-only positive cells (hepatic), lowest cytokeratin 19 (CK19)-only positive cells (cholangiocytic), and highest mean albumin intensity per cell than HepaRG random cocultures and monocultures; however, 80%-84% of HepaRGs remained bipotential (albumin+/CK19+) across all models. The 3T3-J2s also induced higher albumin in HepaRG spheroids than HepaRG-only spheroids. Additionally, although rifampin induced CYP3A4 in HepaRG-MPCCs and HepaRG-CCs, only HepaRG-MPCCs showed the dual omeprazole-mediated CYP1A2/3A4 induction as with PHHs. Lastly, when treated for 6 days with 47 drugs and evaluated for albumin and ATP to make binary hepatotoxicity calls, HepaRG-MPCCs displayed a sensitivity of 54% and specificity of 100% (70%/100% in PHH-MPCCs), whereas HepaRG-CCs misclassified several hepatotoxins. Ultimately, HepaRG-MPCCs could be a more cost-effective and reproducible model than PHHs for executing a tier 1 compound screen.
Collapse
Affiliation(s)
- Brenton R Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Jennifer S Liu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Kimberly R Ballinger
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Salman R Khetani
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523, USA
| |
Collapse
|
12
|
Cruz-Santiago O, Pérez-Maldonado IN, González-Mille DJ, Espinosa-Reyes G, Martínez-Toledo Á, Ilizaliturri-Hernández CA. Nondestructive biomarkers in giant toad (Rhinella marina) to assess the effect of complex mixture of pollutants in Coatzacoalcos River, Mexico. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 82:103558. [PMID: 33307127 DOI: 10.1016/j.etap.2020.103558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 06/12/2023]
Abstract
In this study, we evaluated the usefulness of nondestructive biomarkers approach in giant toads (Rhinella marina). We obtained blood samples and the residual condition index of toads from rural and industrial zones from Coatzacoalcos River, Mexico (COA). In the blood samples, we determined the activity of enzymes, lipid peroxidation, and the presence of cell death (apoptosis). We found that the activity of the enzyme delta-aminolevulinic dehydratase was lower. Still, the glutathione s-transferase activity and the percentage of apoptosis in erythrocytes were higher in the toads of COA than laboratory toads. Meanwhile, some biomarkers in toads showed differences when compared between Industrial and Rural zones. These results and correlations between biomarkers showed how the response changed in the toads living near the industrial zones. We demonstrate that a nondestructive biomarkers approach can be useful in environmental studies with anuran amphibians.
Collapse
Affiliation(s)
- Omar Cruz-Santiago
- Programa Multidisciplinario de Posgrado en Ciencias Ambientales (PMPCA), Agenda Ambiental, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 201, Zona Universitaria, 78210, San Luis Potosí, Mexico
| | - Iván Nelinho Pérez-Maldonado
- Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), CIACyT - Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, Mexico
| | - Donaji Josefina González-Mille
- Cátedras Consejo Nacional de Ciencia y Tecnología (CONACYT), Universidad Autónoma de San Luis Potosí, Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, Mexico
| | - Guillermo Espinosa-Reyes
- Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), CIACyT - Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, Mexico
| | - Ángeles Martínez-Toledo
- Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), CIACyT - Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, Mexico
| | - César Arturo Ilizaliturri-Hernández
- Centro de Investigación Aplicada en Ambiente y Salud (CIAAS), CIACyT - Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, Mexico.
| |
Collapse
|
13
|
Uppugunduri CRS, Muthukumaran J, Robin S, Santos-Silva T, Ansari M. In silico and in vitro investigations on the protein-protein interactions of glutathione S-transferases with mitogen-activated protein kinase 8 and apoptosis signal-regulating kinase 1. J Biomol Struct Dyn 2020; 40:1430-1440. [PMID: 32996404 DOI: 10.1080/07391102.2020.1827036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Cytosolic glutathione S-transferase (GST) enzymes participate in several cellular processes in addition to facilitating glutathione conjugation reactions that eliminate endogenous and exogenous toxic compounds, especially electrophiles. GSTs are thought to interact with various kinases, resulting in the modulation of apoptotic processes and cellular proliferation. The present research used a combination of in silico and in vitro studies to investigate protein-protein interactions between the seven most abundant cytosolic GSTs-GST alpha-1 (GST-A1), GST alpha-2 (GST-A2), GST mu-1 (GST-M1), GST mu-2 (GST-M2), GST mu-5 (GST-M5), GST theta-1 (GST-T1) and GST pi-1 (GST-P1)-and Mitogen-activated protein kinase 8 (MAPK8) and Apoptosis signal-regulating kinase 1 (ASK1). MAPK8 and ASK1 were chosen as this study's protein interaction partners because of their predominant role in electrophile or cytokine-induced stress-mediated apoptosis, inflammation and fibrosis. The highest degree of sequence homology or sequence similarity was observed in two GST subgroups: the GST-A1, GST-A2 and GST-P1 isoforms constituted subgroup1; the GST-M1, GST-M2 and GST-M5 isoforms constituted subgroup 2. The GST-T1 isoform diverged from these isoforms. In silico investigations revealed that GST-M1 showed a significantly higher binding affinity to MAPK8, and its complex was more structurally stable than the other isoforms, in the order GST-M1 > GST-M5 > GST-P1 > GST-A2 > GST-A1 > GST-M2 > GST-T1. Similarly, GST-A1, GST-P1 and GST-T1 actively interacted with ASK1, and their structural stability was also better, in the order GST-T1 > GST-A1 > GST-P1 > GST-A2 > GST-M5 > GST-M1 > GST-M2. To validate in silico results, we performed in vitro crosslinking and mass spectroscopy experiments. Results indicated that GST-M1 interacted with GST-T1 to form heterodimers and confirmed the predicted interaction between GST-M1 and MAPK8.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chakradhara Rao S Uppugunduri
- Onco-Haematology Unit, Department of Paediatrics, Obstetrics and Gynaecology, Geneva University Hospitals, Geneva, Switzerland.,Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jayaraman Muthukumaran
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal.,Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Shannon Robin
- Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Teresa Santos-Silva
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Marc Ansari
- Onco-Haematology Unit, Department of Paediatrics, Obstetrics and Gynaecology, Geneva University Hospitals, Geneva, Switzerland.,Research Platform on Pediatric Onco-Hematology, Department of Paediatrics, Obstetrics and Gynaecology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
14
|
Brys R, Gibson K, Poljak T, Van Der Plas S, Amantini D. Discovery and development of ASK1 inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2020; 59:101-179. [PMID: 32362327 DOI: 10.1016/bs.pmch.2020.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant activation of mitogen-activated protein kinases (MAPKs) like c-Jun N-terminal kinase (JNK) and p38 is an event involved in the pathophysiology of numerous human diseases. The apoptosis signal-regulating kinase 1 (ASK1) is an upstream target that gets activated only under pathological conditions and as such is a promising target for therapeutic intervention. In the first part of this review the molecular mechanisms leading to ASK1 activation and regulation will be described as well as the evidences supporting a pathogenic role for ASK1 in human disease. In the second part, an update on drug discovery efforts towards the discovery and development of ASK1-targeting therapies will be provided.
Collapse
Affiliation(s)
| | - Karl Gibson
- Sandexis Medicinal Chemistry Ltd, Innovation House Discovery ParkSandwich, Kent, United Kingdom
| | | | | | | |
Collapse
|
15
|
Morales-Prieto N, Huertas-Abril PV, López de Lerma N, Pacheco IL, Pérez J, Peinado R, Abril N. Pedro Ximenez sun-dried grape must: a dietary supplement for a healthy longevity. Food Funct 2020; 11:4387-4402. [DOI: 10.1039/d0fo00204f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sun-dried Pedro Ximénez white grapes must (PXM) is a potent antioxidant that regularizes apoptosis, proliferation, and regeneration of the structure and the function of aged mice liver. PXM consumption contributes to a healthy aging process.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Paula V. Huertas-Abril
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | | | - Isabel. L. Pacheco
- Departamento de Anatomía y Anatomía Patológica Comparadas. Facultad de Veterinaria. Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - José Pérez
- Departamento de Anatomía y Anatomía Patológica Comparadas. Facultad de Veterinaria. Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Rafael Peinado
- Departamento de Química Agrícola
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| |
Collapse
|
16
|
Park JG, Aziz N, Cho JY. MKK7, the essential regulator of JNK signaling involved in cancer cell survival: a newly emerging anticancer therapeutic target. Ther Adv Med Oncol 2019; 11:1758835919875574. [PMID: 31579105 PMCID: PMC6759727 DOI: 10.1177/1758835919875574] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/19/2019] [Indexed: 01/02/2023] Open
Abstract
One of the mitogen-activated protein kinases (MAPKs), c-Jun NH2-terminal protein kinase (JNK) plays an important role in regulating cell fate, such as proliferation, differentiation, development, transformation, and apoptosis. Its activity is induced through the interaction of MAPK kinase kinases (MAP3Ks), MAPK kinases (MAP2Ks), and various scaffolding proteins. Because of the importance of the JNK cascade to intracellular bioactivity, many studies have been conducted to reveal its precise intracellular functions and mechanisms, but its regulatory mechanisms remain elusive. In this review, we discuss the molecular characterization, activation process, and physiological functions of mitogen-activated protein kinase kinase 7 (MKK7), the MAP2K that most specifically controls the activity of JNK. Understanding the role of MKK7/JNK signaling in physiological conditions could spark new hypotheses for targeted anticancer therapies.
Collapse
Affiliation(s)
- Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| |
Collapse
|
17
|
Vlach M, Quesnot N, Dubois-Pot-Schneider H, Ribault C, Verres Y, Petitjean K, Rauch C, Morel F, Robin MA, Corlu A, Loyer P. Cytochrome P450 1A1/2, 2B6 and 3A4 HepaRG Cell-Based Biosensors to Monitor Hepatocyte Differentiation, Drug Metabolism and Toxicity. SENSORS 2019; 19:s19102245. [PMID: 31096615 PMCID: PMC6567340 DOI: 10.3390/s19102245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/20/2023]
Abstract
Human hepatoma HepaRG cells express most drug metabolizing enzymes and constitute a pertinent in vitro alternative cell system to primary cultures of human hepatocytes in order to determine drug metabolism and evaluate the toxicity of xenobiotics. In this work, we established novel transgenic HepaRG cells transduced with lentiviruses encoding the reporter green fluorescent protein (GFP) transcriptionally regulated by promoter sequences of cytochromes P450 (CYP) 1A1/2, 2B6 and 3A4 genes. Here, we demonstrated that GFP-biosensor transgenes shared similar expression patterns with the corresponding endogenous CYP genes during proliferation and differentiation in HepaRG cells. Interestingly, differentiated hepatocyte-like HepaRG cells expressed GFP at higher levels than cholangiocyte-like cells. Despite weaker inductions of GFP expression compared to the strong increases in mRNA levels of endogenous genes, we also demonstrated that the biosensor transgenes were induced by prototypical drug inducers benzo(a)pyrene and phenobarbital. In addition, we used the differentiated biosensor HepaRG cells to evidence that pesticide mancozeb triggered selective cytotoxicity of hepatocyte-like cells. Our data demonstrate that these new biosensor HepaRG cells have potential applications in the field of chemicals safety evaluation and the assessment of drug hepatotoxicity.
Collapse
Affiliation(s)
- Manuel Vlach
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Nicolas Quesnot
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | | | - Catherine Ribault
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Yann Verres
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Kilian Petitjean
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Claudine Rauch
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Fabrice Morel
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Marie-Anne Robin
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Anne Corlu
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Pascal Loyer
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
- Correspondence: ; Tel.: +33-(0)223233873; Fax: +33-(0)299540137
| |
Collapse
|
18
|
Tascher G, Burban A, Camus S, Plumel M, Chanon S, Le Guevel R, Shevchenko V, Van Dorsselaer A, Lefai E, Guguen-Guillouzo C, Bertile F. In-Depth Proteome Analysis Highlights HepaRG Cells as a Versatile Cell System Surrogate for Primary Human Hepatocytes. Cells 2019; 8:E192. [PMID: 30795634 PMCID: PMC6406872 DOI: 10.3390/cells8020192] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Of the hepatic cell lines developed for in vitro studies of hepatic functions as alternatives to primary human hepatocytes, many have lost major liver-like functions, but not HepaRG cells. The increasing use of the latter worldwide raises the need for establishing the reference functional status of early biobanked HepaRG cells. Using deep proteome and secretome analyses, the levels of master regulators of the hepatic phenotype and of the structural elements ensuring biliary polarity were found to be close to those in primary hepatocytes. HepaRG cells proved to be highly differentiated, with functional mitochondria, hepatokine secretion abilities, and an adequate response to insulin. Among differences between primary human hepatocytes and HepaRG cells, the factors that possibly support HepaRG transdifferentiation properties are discussed. The HepaRG cell system thus appears as a robust surrogate for primary hepatocytes, which is versatile enough to study not only xenobiotic detoxification, but also the control of hepatic energy metabolism, secretory function and disease-related mechanisms.
Collapse
Affiliation(s)
- Georg Tascher
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
- Institute of Biochemistry II, Goethe University Hospital, D-60590 Frankfurt am Main, Germany.
| | - Audrey Burban
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
| | - Sandrine Camus
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Marine Plumel
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Stéphanie Chanon
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Remy Le Guevel
- ImPACcell platform, Biosit, Université de Rennes 1, F-35043 Rennes, France.
| | - Valery Shevchenko
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Alain Van Dorsselaer
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Etienne Lefai
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Christiane Guguen-Guillouzo
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Fabrice Bertile
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| |
Collapse
|
19
|
Vilas-Boas V, Cooreman A, Gijbels E, Van Campenhout R, Gustafson E, Ballet S, Annaert P, Cogliati B, Vinken M. Primary hepatocytes and their cultures for the testing of drug-induced liver injury. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 85:1-30. [PMID: 31307583 DOI: 10.1016/bs.apha.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury is a major reason for discontinuation of drug development and withdrawal of drugs from the market. Intensive efforts in the last decades have focused on the establishment and finetuning of liver-based in vitro models for reliable prediction of hepatotoxicity triggered by drug candidates. Of those, primary hepatocytes and their cultures still are considered the gold standard, as they provide an acceptable reflection of the hepatic in vivo situation. Nevertheless, these in vitro systems cope with gradual deterioration of the differentiated morphological and functional phenotype. The present paper gives an overview of traditional and more recently introduced strategies to counteract this dedifferentiation process in an attempt to set up culture models that can be used for long-term testing purposes. The relevance and applicability of such optimized cultures of primary hepatocytes for the testing of drug-induced cholestatic liver injury is demonstrated.
Collapse
Affiliation(s)
- Vânia Vilas-Boas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Axelle Cooreman
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Gijbels
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emma Gustafson
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
20
|
Pajares MA, Pérez-Sala D. Mammalian Sulfur Amino Acid Metabolism: A Nexus Between Redox Regulation, Nutrition, Epigenetics, and Detoxification. Antioxid Redox Signal 2018; 29:408-452. [PMID: 29186975 DOI: 10.1089/ars.2017.7237] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Transsulfuration allows conversion of methionine into cysteine using homocysteine (Hcy) as an intermediate. This pathway produces S-adenosylmethionine (AdoMet), a key metabolite for cell function, and provides 50% of the cysteine needed for hepatic glutathione synthesis. The route requires the intake of essential nutrients (e.g., methionine and vitamins) and is regulated by their availability. Transsulfuration presents multiple interconnections with epigenetics, adenosine triphosphate (ATP), and glutathione synthesis, polyol and pentose phosphate pathways, and detoxification that rely mostly in the exchange of substrates or products. Major hepatic diseases, rare diseases, and sensorineural disorders, among others that concur with oxidative stress, present impaired transsulfuration. Recent Advances: In contrast to the classical view, a nuclear branch of the pathway, potentiated under oxidative stress, is emerging. Several transsulfuration proteins regulate gene expression, suggesting moonlighting activities. In addition, abnormalities in Hcy metabolism link nutrition and hearing loss. CRITICAL ISSUES Knowledge about the crossregulation between pathways is mostly limited to the hepatic availability/removal of substrates and inhibitors. However, advances regarding protein-protein interactions involving oncogenes, identification of several post-translational modifications (PTMs), and putative moonlighting activities expand the potential impact of transsulfuration beyond methylations and Hcy. FUTURE DIRECTIONS Increasing the knowledge on transsulfuration outside the liver, understanding the protein-protein interaction networks involving these enzymes, the functional role of their PTMs, or the mechanisms controlling their nucleocytoplasmic shuttling may provide further insights into the pathophysiological implications of this pathway, allowing design of new therapeutic interventions. Antioxid. Redox Signal. 29, 408-452.
Collapse
Affiliation(s)
- María A Pajares
- 1 Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas (CSIC) , Madrid, Spain .,2 Molecular Hepatology Group, Instituto de Investigación Sanitaria La Paz (IdiPAZ) , Madrid, Spain
| | - Dolores Pérez-Sala
- 1 Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas (CSIC) , Madrid, Spain
| |
Collapse
|
21
|
Burban A, Sharanek A, Guguen-Guillouzo C, Guillouzo A. Endoplasmic reticulum stress precedes oxidative stress in antibiotic-induced cholestasis and cytotoxicity in human hepatocytes. Free Radic Biol Med 2018; 115:166-178. [PMID: 29191461 DOI: 10.1016/j.freeradbiomed.2017.11.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/11/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) stress has been associated with various drug-induced liver lesions but its participation in drug-induced cholestasis remains unclear. We first aimed at analyzing liver damage caused by various hepatotoxic antibiotics, including three penicillinase-resistant antibiotics (PRAs), i.e. flucloxacillin, cloxacillin and nafcillin, as well as trovafloxacin, levofloxacin and erythromycin, using human differentiated HepaRG cells and primary hepatocytes. All these antibiotics caused early cholestatic effects typified by bile canaliculi dilatation and reduced bile acid efflux within 2h and dose-dependent enhanced caspase-3 activity within 24h. PRAs induced the highest cholestatic effects at non cytotoxic concentrations. Then, molecular events involved in these lesions were analyzed. Early accumulation of misfolded proteins revealed by thioflavin-T fluorescence and associated with phosphorylation of the unfolded protein response sensors, eIF2α and/or IRE1α, was evidenced with all tested hepatotoxic antibiotics. Inhibition of ER stress markedly restored bile acid efflux and prevented bile canaliculi dilatation. Downstream of ER stress, ROS were also generated with high antibiotic concentrations. The protective HSP27-PI3K-AKT signaling pathway was activated only in PRA-treated cells and its inhibition increased ROS production and aggravated caspase-3 activity. Overall, our results demonstrate that (i) various antibiotics reported to cause cholestasis and hepatocellular injury in the clinic can also induce such effects in in vitro human hepatocytes; (ii) PRAs cause the strongest cholestatic effects in the absence of cytotoxicity; (iii) cholestatic features occur early through ER stress; (iv) cytotoxic lesions are observed later through ER stress-mediated ROS generation; and (v) activation of the HSP27-PI3K-AKT pathway protects from cytotoxic damage induced by PRAs only.
Collapse
Affiliation(s)
- Audrey Burban
- INSERM U991/1241, Numecan, Rennes, France; Rennes 1 University, Rennes, France
| | - Ahmad Sharanek
- INSERM U991/1241, Numecan, Rennes, France; Rennes 1 University, Rennes, France
| | | | - André Guillouzo
- INSERM U991/1241, Numecan, Rennes, France; Rennes 1 University, Rennes, France.
| |
Collapse
|
22
|
Differential sensitivity of metabolically competent and non-competent HepaRG cells to apoptosis induced by diclofenac combined or not with TNF-α. Toxicol Lett 2016; 258:71-86. [DOI: 10.1016/j.toxlet.2016.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 01/20/2023]
|
23
|
Sánchez-Gómez FJ, Díez-Dacal B, García-Martín E, Agúndez JAG, Pajares MA, Pérez-Sala D. Detoxifying Enzymes at the Cross-Roads of Inflammation, Oxidative Stress, and Drug Hypersensitivity: Role of Glutathione Transferase P1-1 and Aldose Reductase. Front Pharmacol 2016; 7:237. [PMID: 27540362 PMCID: PMC4973429 DOI: 10.3389/fphar.2016.00237] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/21/2016] [Indexed: 01/01/2023] Open
Abstract
Phase I and II enzymes are involved in the metabolism of endogenous reactive compounds as well as xenobiotics, including toxicants and drugs. Genotyping studies have established several drug metabolizing enzymes as markers for risk of drug hypersensitivity. However, other candidates are emerging that are involved in drug metabolism but also in the generation of danger or costimulatory signals. Enzymes such as aldo-keto reductases (AKR) and glutathione transferases (GST) metabolize prostaglandins and reactive aldehydes with proinflammatory activity, as well as drugs and/or their reactive metabolites. In addition, their metabolic activity can have important consequences for the cellular redox status, and impacts the inflammatory response as well as the balance of inflammatory mediators, which can modulate epigenetic factors and cooperate or interfere with drug-adduct formation. These enzymes are, in turn, targets for covalent modification and regulation by oxidative stress, inflammatory mediators, and drugs. Therefore, they constitute a platform for a complex set of interactions involving drug metabolism, protein haptenation, modulation of the inflammatory response, and/or generation of danger signals with implications in drug hypersensitivity reactions. Moreover, increasing evidence supports their involvement in allergic processes. Here, we will focus on GSTP1-1 and aldose reductase (AKR1B1) and provide a perspective for their involvement in drug hypersensitivity.
Collapse
Affiliation(s)
- Francisco J Sánchez-Gómez
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | - Beatriz Díez-Dacal
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | | | - José A G Agúndez
- Department of Pharmacology, University of Extremadura Cáceres, Spain
| | - María A Pajares
- Instituto de Investigaciones Biomédicas Alberto Sols (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), and Grupo de Hepatología Molecular, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ) Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| |
Collapse
|
24
|
Pajaud J, Ribault C, Ben Mosbah I, Rauch C, Henderson C, Bellaud P, Aninat C, Loyer P, Morel F, Corlu A. Glutathione transferases P1/P2 regulate the timing of signaling pathway activations and cell cycle progression during mouse liver regeneration. Cell Death Dis 2015; 6:e1598. [PMID: 25590808 PMCID: PMC4669760 DOI: 10.1038/cddis.2014.562] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/01/2023]
Abstract
Glutathione transferases (GST) are phase II enzymes catalyzing the detoxification of endogenous noxious compounds and xenobiotics. They also regulate phosphorylation activities of MAPKinases in a catalytic-independent manner. Previous studies have demonstrated the regulation of JNK-dependent pathway by GSTP1/2. Considering the crucial role of JNK in the early steps of the hepatocyte cell cycle, we sought to determine whether GSTP1/2 were essential for hepatocyte proliferation following partial hepatectomy (PH). Using a conventional double knockout mouse model for the Gstp1 and Gstp2 genes, we found that the lack of GSTP1/P2 reduced the rate of DNA replication and mitotic index during the first wave of hepatocyte proliferation. The lowered proliferation was associated with the decrease in TNFalpha and IL-6 plasma concentrations, reduced hepatic HGF expression and delayed and/or altered activation of STAT3, JNK and ERK1/2 signaling pathways. In addition, the expression and/or activation of cell cycle regulators such as Cyclin D1, CDK4, E2F1 and MCM7 was postponed demonstrating that the absence of GSTP1/2 delayed the entry into and progression through the G1 phase of the cell cycle and impaired the synchrony of proliferation in hepatocytes following PH. Furthermore, while JNK and its downstream targets c-Jun and ATF2 were activated during the early steps of the liver regeneration in wild-type animals, the constitutively active JNK found in the quiescent liver of Gstp1/2 knockout mice underwent a decrease in its activity after PH. Transient induction of antioxidant enzymes and nitric oxide synthase were also delayed or repressed during the regenerative response. Altogether our results demonstrate that GSTP1/2 are a critical regulators of hepatocyte proliferation in the initial phases of liver regeneration.
Collapse
Affiliation(s)
- J Pajaud
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - C Ribault
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - I Ben Mosbah
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - C Rauch
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - C Henderson
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - P Bellaud
- Université de Rennes 1, Faculté de Médecine, Rennes, France
- Plateforme Histopathologie H2P2, Biosit, Biogenouest, Université de Rennes 1, Rennes, France
| | - C Aninat
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - P Loyer
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - F Morel
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| | - A Corlu
- Inserm, UMR 991, Liver, Metabolisms and Cancer, CHU Pontchaillou, Rennes, France
- Université de Rennes 1, Faculté de Médecine, Rennes, France
| |
Collapse
|
25
|
Corlu A, Loyer P. Culture Conditions Promoting Hepatocyte Proliferation and Cell Cycle Synchronization. Methods Mol Biol 2015; 1250:27-51. [PMID: 26272133 DOI: 10.1007/978-1-4939-2074-7_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The liver overcomes damages induced by harmful substances or viral infections and allows the use of extended resection in human therapy through its remarkable ability to regenerate. The regeneration process relies on the massive proliferation of differentiated hepatocytes that exit quiescence and undergo a limited number of cell cycles to restore the hepatic mass. Many discoveries on the regulation of hepatocyte proliferation have benefited from the use of in vitro models of cultures of primary hepatocytes as well as hepatoma cells as opposed to data obtained from in vivo models of liver regeneration, such as following partial hepatectomy in rodents. In this chapter, the most pertinent in vitro models used to promote the proliferation of hepatocytes and technical procedures to synchronize their progression throughout the cell cycle are presented with the goal to investigate the regulation of the hepatocyte cell cycle and the molecular pathways regulating liver regeneration.
Collapse
Affiliation(s)
- Anne Corlu
- Inserm, UMR 991, Liver, Metabolisms and Cancer, Hôpital Pontchaillou, University of Rennes 1, Rennes Cedex, 35033, France
| | | |
Collapse
|
26
|
Barouti G, Jarnouen K, Cammas-Marion S, Loyer P, Guillaume SM. Polyhydroxyalkanoate-based amphiphilic diblock copolymers as original biocompatible nanovectors. Polym Chem 2015. [DOI: 10.1039/c5py00831j] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticles derived from poly(β-malic acid)-b-poly(3-hydroxybutyrate) (PMLA-b-PHB) copolymers revealed no cytotoxicity towards HepaRG and SK-MEL-28 cells.
Collapse
Affiliation(s)
- Ghislaine Barouti
- Institut des Sciences Chimiques de Rennes
- UMR 6226 CNRS – Université de Rennes 1
- F-35042 Rennes Cedex
- France
| | | | - Sandrine Cammas-Marion
- Ecole Nationale Supérieure de Chimie de Rennes – Institut des Sciences Chimiques de Rennes
- UMR 6226 CNRS – Université de Rennes 1
- F-35708 Rennes Cedex
- France
| | - Pascal Loyer
- INSERM
- UMR991
- Liver
- Metabolisms and Cancer
- CHU Pontchaillou
| | - Sophie M. Guillaume
- Institut des Sciences Chimiques de Rennes
- UMR 6226 CNRS – Université de Rennes 1
- F-35042 Rennes Cedex
- France
| |
Collapse
|
27
|
Collin A, Hardonnière K, Chevanne M, Vuillemin J, Podechard N, Burel A, Dimanche-Boitrel MT, Lagadic-Gossmann D, Sergent O. Cooperative interaction of benzo[a]pyrene and ethanol on plasma membrane remodeling is responsible for enhanced oxidative stress and cell death in primary rat hepatocytes. Free Radic Biol Med 2014; 72:11-22. [PMID: 24681337 DOI: 10.1016/j.freeradbiomed.2014.03.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/14/2014] [Accepted: 03/19/2014] [Indexed: 10/25/2022]
Abstract
Several epidemiologic studies have shown an interactive effect of heavy smoking and heavy alcohol drinking on the development of hepatocellular carcinoma. It has also been recently described that chronic hepatocyte death can trigger excessive compensatory proliferation resulting later in the formation of tumors in mouse liver. As we previously demonstrated that both benzo[a]pyrene (B[a]P), an environmental agent found in cigarette smoke, and ethanol possess similar targets, especially oxidative stress, to trigger death of liver cells, we decided to study here the cellular and molecular mechanisms of the effects of B[a]P/ethanol coexposure on cell death. After an 18-h incubation with 100nM B[a]P, primary rat hepatocytes were supplemented with 50mM ethanol for 5 or 8h. B[a]P/ethanol coexposure led to a greater apoptotic cell death that could be linked to an increase in lipid peroxidation. Plasma membrane remodeling, as depicted by membrane fluidity elevation and physicochemical alterations in lipid rafts, appeared to play a key role, because both toxicants acted with specific complementary effects. Membrane remodeling was shown to induce an accumulation of lysosomes leading to an important increase in low-molecular-weight iron cellular content. Finally, ethanol metabolism, but not that of B[a]P, by providing reactive oxygen species, induced the ultimate toxic process. Indeed, in lysosomes, ethanol promoted the Fenton reaction, lipid peroxidation, and membrane permeabilization, thereby triggering cell death. To conclude, B[a]P exposure, by depleting hepatocyte membrane cholesterol content, would constitute a favorable ground for a later toxic insult such as ethanol intoxication. Membrane stabilization of both plasma membrane and lysosomes might be a potential target for further investigation considering cytoprotective strategies.
Collapse
Affiliation(s)
- Aurore Collin
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Kevin Hardonnière
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Martine Chevanne
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Julie Vuillemin
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Normand Podechard
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Agnès Burel
- Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Marie-Thérèse Dimanche-Boitrel
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France
| | - Dominique Lagadic-Gossmann
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France.
| | - Odile Sergent
- UMR INSERM 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, and 35043 Rennes Cédex, France; Biosit UMS3080, Université de Rennes 1, 35043 Rennes Cédex, France.
| |
Collapse
|
28
|
Stocco G, Pelin M, Franca R, De Iudicibus S, Cuzzoni E, Favretto D, Martelossi S, Ventura A, Decorti G. Pharmacogenetics of azathioprine in inflammatory bowel disease: a role for glutathione-S-transferase? World J Gastroenterol 2014; 20:3534-3541. [PMID: 24707136 PMCID: PMC3974520 DOI: 10.3748/wjg.v20.i13.3534] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/30/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023] Open
Abstract
Azathioprine is a purine antimetabolite drug commonly used to treat inflammatory bowel disease (IBD). In vivo it is active after reaction with reduced glutathione (GSH) and conversion to mercaptopurine. Although this reaction may occur spontaneously, the presence of isoforms M and A of the enzyme glutathione-S-transferase (GST) may increase its speed. Indeed, in pediatric patients with IBD, deletion of GST-M1, which determines reduced enzymatic activity, was recently associated with reduced sensitivity to azathioprine and reduced production of azathioprine active metabolites. In addition to increase the activation of azathioprine to mercaptopurine, GSTs may contribute to azathioprine effects even by modulating GSH consumption, oxidative stress and apoptosis. Therefore, genetic polymorphisms in genes for GSTs may be useful to predict response to azathioprine even if more in vitro and clinical validation studies are needed.
Collapse
|
29
|
Cellular impact of combinations of endosulfan, atrazine, and chlorpyrifos on human primary hepatocytes and HepaRG cells after short and chronic exposures. Cell Biol Toxicol 2013; 30:17-29. [DOI: 10.1007/s10565-013-9266-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 12/02/2013] [Indexed: 12/31/2022]
|
30
|
Primary hepatocytes and their cultures in liver apoptosis research. Arch Toxicol 2013; 88:199-212. [PMID: 24013573 DOI: 10.1007/s00204-013-1123-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/29/2013] [Indexed: 01/18/2023]
Abstract
Apoptosis not only plays a key role in physiological demise of defunct hepatocytes, but is also associated with a plethora of acute and chronic liver diseases as well as with hepatotoxicity. The present paper focuses on the modelling of this mode of programmed cell death in primary hepatocyte cultures. Particular attention is paid to the activation of spontaneous apoptosis during the isolation of hepatocytes from the liver, its progressive manifestation upon the subsequent establishment of cell cultures and simultaneously to strategies to counteract this deleterious process. In addition, currently applied approaches to experimentally induce controlled apoptosis in this in vitro setting for mechanistic research purposes and thereby its detection using relevant biomarkers are reviewed.
Collapse
|
31
|
Klaus A, Zorman S, Berthier A, Polge C, Ramirez S, Michelland S, Sève M, Vertommen D, Rider M, Lentze N, Auerbach D, Schlattner U. Glutathione S-transferases interact with AMP-activated protein kinase: evidence for S-glutathionylation and activation in vitro. PLoS One 2013; 8:e62497. [PMID: 23741294 PMCID: PMC3669356 DOI: 10.1371/journal.pone.0062497] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/22/2013] [Indexed: 11/18/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a cellular and whole body energy sensor with manifold functions in regulating energy homeostasis, cell morphology and proliferation in health and disease. Here we apply multiple, complementary in vitro and in vivo interaction assays to identify several isoforms of glutathione S-transferase (GST) as direct AMPK binding partners: Pi-family member rat GSTP1 and Mu-family members rat GSTM1, as well as Schistosoma japonicum GST. GST/AMPK interaction is direct and involves the N-terminal domain of the AMPK β-subunit. Complex formation of the mammalian GSTP1 and -M1 with AMPK leads to their enzymatic activation and in turn facilitates glutathionylation and activation of AMPK in vitro. GST-facilitated S-glutathionylation of AMPK may be involved in rapid, full activation of the kinase under mildly oxidative physiological conditions.
Collapse
Affiliation(s)
- Anna Klaus
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
| | - Sarah Zorman
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
| | - Alexandre Berthier
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
| | - Cécile Polge
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
| | - Sacnicte Ramirez
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
| | - Sylvie Michelland
- CRI-Inserm, Institut Albert Bonniot, Grenoble, France
- Centre Hospitalier Universitaire Grenoble, Plate-forme Protéomique Prométhée, Institut de Biologie et Pathologie, Grenoble, France
| | - Michel Sève
- CRI-Inserm, Institut Albert Bonniot, Grenoble, France
- Centre Hospitalier Universitaire Grenoble, Plate-forme Protéomique Prométhée, Institut de Biologie et Pathologie, Grenoble, France
| | - Didier Vertommen
- Université Catholique de Louvain, Faculty of Medicine and de Duve Institute, Brussels, Belgium
| | - Mark Rider
- Université Catholique de Louvain, Faculty of Medicine and de Duve Institute, Brussels, Belgium
| | | | | | - Uwe Schlattner
- Université Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
- Inserm, Grenoble, France
- * E-mail:
| |
Collapse
|
32
|
Corlu A, Loyer P. Regulation of the g1/s transition in hepatocytes: involvement of the cyclin-dependent kinase cdk1 in the DNA replication. Int J Hepatol 2012; 2012:689324. [PMID: 23091735 PMCID: PMC3471441 DOI: 10.1155/2012/689324] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022] Open
Abstract
A singular feature of adult differentiated hepatocytes is their capacity to proliferate allowing liver regeneration. This review emphasizes the literature published over the last 20 years that established the most important pathways regulating the hepatocyte cell cycle. Our article also aimed at illustrating that many discoveries in this field benefited from the combined use of in vivo models of liver regeneration and in vitro models of primary cultures of human and rodent hepatocytes. Using these models, our laboratory has contributed to decipher the different steps of the progression into the G1 phase and the commitment to S phase of proliferating hepatocytes. We identified the mitogen dependent restriction point located at the two-thirds of the G1 phase and the concomitant expression and activation of both Cdk1 and Cdk2 at the G1/S transition. Furthermore, we demonstrated that these two Cdks contribute to the DNA replication. Finally, we provided strong evidences that Cdk1 expression and activation is correlated to extracellular matrix degradation upon stimulation by the pro-inflammatory cytokine TNFα leading to the identification of a new signaling pathway regulating Cdk1 expression at the G1/S transition. It also further confirms the well-orchestrated regulation of liver regeneration via multiple extracellular signals and pathways.
Collapse
Affiliation(s)
- Anne Corlu
- Inserm UMR S 991, Foie Métabolismes et Cancer, Université de Rennes 1, Hôpital Pontchaillou, 35033 Rennes Cedex, France
| | - Pascal Loyer
- Inserm UMR S 991, Foie Métabolismes et Cancer, Université de Rennes 1, Hôpital Pontchaillou, 35033 Rennes Cedex, France
| |
Collapse
|
33
|
Wu W, Peden D, Diaz-Sanchez D. Role of GSTM1 in resistance to lung inflammation. Free Radic Biol Med 2012; 53:721-9. [PMID: 22683820 PMCID: PMC3418458 DOI: 10.1016/j.freeradbiomed.2012.05.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 01/04/2023]
Abstract
Lung inflammation resulting from oxidant/antioxidant imbalance is a common feature of many lung diseases. In particular, the role of enzymes regulated by the NF-E2-related factor 2 transcription factor has recently received increased attention. Among these antioxidant genes, glutathione S-transferase Mu 1 (GSTM1) has been most extensively characterized because it has a null polymorphism that is highly prevalent in the population and associated with increased risk of inflammatory lung diseases. Present evidence suggests that GSTM1 acts through interactions with other genes and environmental factors, especially air pollutants. Here, we review GSTM1 gene expression and regulation and summarize the findings from epidemiological, clinical, animal, and in vitro studies on the role played by GSTM1 in lung inflammation. We discuss limitations in the existing knowledge base and future perspectives and evaluate the potential of pharmacologic and genetic manipulation of the GSTM1 gene to modulate pulmonary inflammatory responses.
Collapse
Affiliation(s)
- Weidong Wu
- Department of Pediatrics, Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC 7599, USA.
| | | | | |
Collapse
|
34
|
Optimization of the HepaRG cell model for drug metabolism and toxicity studies. Toxicol In Vitro 2012; 26:1278-85. [PMID: 22643240 DOI: 10.1016/j.tiv.2012.05.008] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 05/15/2012] [Accepted: 05/18/2012] [Indexed: 01/08/2023]
Abstract
The HepaRG cell line is the first human cell line able to differentiate in vitro into mature hepatocyte-like cells. Our main objective within the framework of the EEC-LIINTOP project was to optimize the use of this cell line for drug metabolism and toxicity studies, especially after repeat treatments. The main results showed that differentiated HepaRG cells: (i) retained their drug metabolism capacity (major CYPs, phase 2 enzymes, transporters and nuclear receptors) and responsiveness to prototypical inducers at relatively stable levels for several weeks at confluence. The levels of several functions, including some CYPs such as CYP3A4, were dependent on the addition of dimethyl sulfoxide in the culture medium; (ii) sustained the different types of chemical-induced hepatotoxicity, including steatosis, phospholipidosis and cholestasis, after acute and/or repeat treatment with reference drugs. In particular, drug-induced vesicular steatosis was demonstrated in vitro for the first time. In conclusion, our results from the LIINTOP project, together with other studies reported concomitantly or more recently in the literature, support the conclusion that the metabolically competent human HepaRG cells represent a surrogate to primary human hepatocytes for investigating drug metabolism parameters and both acute and chronic effects of xenobiotics in human liver.
Collapse
|
35
|
Gerets HHJ, Tilmant K, Gerin B, Chanteux H, Depelchin BO, Dhalluin S, Atienzar FA. Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biol Toxicol 2012; 28:69-87. [PMID: 22258563 PMCID: PMC3303072 DOI: 10.1007/s10565-011-9208-4] [Citation(s) in RCA: 503] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 12/12/2011] [Indexed: 12/15/2022]
Abstract
In the pharmaceutical industry, improving the early detection of drug-induced hepatotoxicity is essential as it is one of the most important reasons for attrition of candidate drugs during the later stages of drug development. The first objective of this study was to better characterize different cellular models (i.e., HepG2, HepaRG cells, and fresh primary human hepatocytes) at the gene expression level and analyze their metabolic cytochrome P450 capabilities. The cellular models were exposed to three different CYP450 inducers; beta-naphthoflavone (BNF), phenobarbital (PB), and rifampicin (RIF). HepG2 cells responded very weakly to the different inducers at the gene expression level, and this translated generally into low CYP450 activities in the induced cells compared with the control cells. On the contrary, HepaRG cells and the three human donors were inducible after exposure to BNF, PB, and RIF according to gene expression responses and CYP450 activities. Consequently, HepaRG cells could be used in screening as a substitute and/or in complement to primary hepatocytes for CYP induction studies. The second objective was to investigate the predictivity of the different cellular models to detect hepatotoxins (16 hepatotoxic and 5 nonhepatotoxic compounds). Specificity was 100% with the different cellular models tested. Cryopreserved human hepatocytes gave the highest sensitivity, ranging from 31% to 44% (depending on the donor), followed by lower sensitivity (13%) for HepaRG and HepG2 cells (6.3%). Overall, none of the models under study gave desirable sensitivities (80-100%). Consequently, a high metabolic capacity and CYP inducibility in cell lines does not necessarily correlate with a high sensitivity for the detection of hepatotoxic drugs. Further investigations are necessary to compare different cellular models and determine those that are best suited for the detection of hepatotoxic compounds.
Collapse
|
36
|
Regulation of signal transduction by glutathione transferases. Int J Hepatol 2012; 2012:137676. [PMID: 23094162 PMCID: PMC3474235 DOI: 10.1155/2012/137676] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/13/2012] [Accepted: 09/13/2012] [Indexed: 12/11/2022] Open
Abstract
Glutathione transferases (GST) are essentially known as enzymes that catalyse the conjugation of glutathione to various electrophilic compounds such as chemical carcinogens, environmental pollutants, and antitumor agents. However, this protein family is also involved in the metabolism of endogenous compounds which play critical roles in the regulation of signaling pathways. For example, the lipid peroxidation product 4-hydroxynonenal (4-HNE) and the prostaglandin 15-deoxy-Δ12,14-prostaglandin J(2) (15d-PGJ(2)) are metabolized by GSTs and these compounds are known to influence the activity of transcription factors and protein kinases involved in stress response, proliferation, differentiation, or apoptosis. Furthermore, several studies have demonstrated that GSTs are able to interact with different protein partners such as mitogen activated protein kinases (i.e., c-jun N-terminal kinase (JNK) and apoptosis signal-regulating kinase 1 (ASK1)) which are also involved in cell signaling. New functions of GSTs, including S-glutathionylation of proteins by GSTs and ability to be a nitric oxide (NO) carrier have also been described. Taken together, these observations strongly suggest that GST might play a crucial role during normal or cancer cells proliferation or apoptosis.
Collapse
|
37
|
Combined Stimulation with the Tumor Necrosis Factor α and the Epidermal Growth Factor Promotes the Proliferation of Hepatocytes in Rat Liver Cultured Slices. Int J Hepatol 2012; 2012:785786. [PMID: 23119170 PMCID: PMC3480011 DOI: 10.1155/2012/785786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/06/2012] [Accepted: 09/07/2012] [Indexed: 11/17/2022] Open
Abstract
The culture liver slices are mainly used to investigate drug metabolism and xenobiotic-mediated liver injuries while apoptosis and proliferation remain unexplored in this culture model. Here, we show a transient increase in LDH release and caspase activities indicating an ischemic injury during the slicing procedure. Then, caspase activities decrease and remain low in cultured slices demonstrating a low level of apoptosis. The slicing procedure is also associated with the G0/G1 transition of hepatocytes demonstrated by the activation of stress and proliferation signalling pathways including the ERK1/2 and JNK1/2/3 MAPKinases and the transient upregulation of c-fos. The cells further progress up to mid-G1 phase as indicated by the sequential induction of c-myc and p53 mRNA levels after the slicing procedure and at 24 h of culture, respectively. The stimulation by epidermal growth factor induces the ERK1/2 phosphorylation but fails to activate expression of late G1 and S phase markers such as cyclin D1 and Cdk1 indicating that hepatocytes are arrested in mid-G1 phase of the cell cycle. However, we found that combined stimulation by the proinflammatory cytokine tumor necrosis factor α and the epidermal growth factor promotes the commitment to DNA replication as observed in vivo during the liver regeneration.
Collapse
|
38
|
Aliche-Djoudi F, Podechard N, Chevanne M, Nourissat P, Catheline D, Legrand P, Dimanche-Boitrel MT, Lagadic-Gossmann D, Sergent O. Physical and chemical modulation of lipid rafts by a dietary n-3 polyunsaturated fatty acid increases ethanol-induced oxidative stress. Free Radic Biol Med 2011; 51:2018-30. [PMID: 21945097 DOI: 10.1016/j.freeradbiomed.2011.08.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/25/2011] [Accepted: 08/25/2011] [Indexed: 12/28/2022]
Abstract
Dietary n-3 polyunsaturated fatty acids (n-3 PUFAs) have been reported to modulate lipid raft-dependent signaling, but not yet lipid raft-dependent oxidative stress. Previously, we have shown that ethanol-induced membrane remodeling, i.e., an increase in membrane fluidity and alterations in physical and biochemical properties of lipid rafts, participated in the development of oxidative stress. Thus, we decided to study n-3 PUFA effects in this context, by pretreating hepatocytes with eicosapentaenoic acid (EPA), a long-chain n-3 PUFA, before addition of ethanol. EPA was found to increase ethanol-induced oxidative stress through membrane remodeling. Addition of EPA resulted in a marked increase in lipid raft aggregation compared to ethanol alone. In addition, membrane fluidity of lipid rafts was markedly enhanced. Interestingly, EPA was found to preferentially incorporate into nonraft membrane regions, leading to raft cholesterol increase. Lipid raft aggregation by EPA enhanced phospholipase Cγ translocation into these microdomains. Finally, phospholipase Cγ was shown to participate in the potentiation of oxidative stress by promoting lysosome accumulation, a major source of low-molecular-weight iron. To conclude, the ability of EPA to modify lipid raft physical and chemical properties plays a key role in the enhancement, by this dietary n-3 PUFA, of ethanol-induced oxidative stress.
Collapse
Affiliation(s)
- Fatiha Aliche-Djoudi
- EA 4427 SeRAIC/IRSET, IFR 140, UFR des Sciences Pharmaceutiques et Biologiques, Université de Rennes 1, Rennes Cédex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Glutathione transferase (GST) kappa, also named mitochondrial GST, is a very ancient protein family with orthologs in bacteria and eukaryotes. Both the structure and the subcellular localization of GSTK1-1, in mitochondria and peroxisomes, make this enzyme distinct from cytosolic GSTs. Rodent and human GSTK1 exhibit activity towards a number of model GST substrates and, in Caenorhabditis elegans, this enzyme may be involved in energy and lipid metabolism, two functions related to mitochondria and peroxisomes. Interestingly, GST kappa is also a key regulator of adiponectin biosynthesis and multimerization suggesting that it might function as a chaperone to facilitate correct folding and assembly of proteins. Since adiponectin expression has been correlated with insulin resistance, obesity and diabetes, GSTK1 expression level which is negatively correlated with obesity in mice and human adipose tissues may be an important factor in these metabolic disorders. Furthermore, a polymorphism in the hGSTK1 promoter has been associated with insulin secretion and fat deposition.
Collapse
Affiliation(s)
- Fabrice Morel
- INSERM UMR991, Université de Rennes 1, F-35033 Rennes, France.
| | | |
Collapse
|
40
|
Abstract
In vitro hepatocyte models represent very useful systems in both fundamental research and various application areas. Primary hepatocytes appear as the closest model for the liver in vivo. However, they are phenotypically unstable, have a limited life span and in addition, exhibit large interdonor variability when of human origin. Hepatoma cell lines appear as an alternative but only the HepaRG cell line exhibits various functions, including major cytochrome P450 activities, at levels close to those found in primary hepatocytes. In vitro hepatocyte models have brought a substantial contribution to the understanding of the biochemistry, physiology, and cell biology of the normal and diseased liver and in various application domains such as xenobiotic metabolism and toxicity, virology, parasitology, and more generally cell therapies. In the future, new well-differentiated hepatocyte cell lines derived from tumors or from either embryonic or adult stem cells might be expected and although hepatocytes will continue to be used in various fields, these in vitro liver models should allow marked advances, especially in cell-based therapies and predictive and mechanistic hepatotoxicity of new drugs and other chemicals. All models will benefit from new developments in throughput screening based on cell chips coupled with high-content imaging and in toxicogenomics technologies.
Collapse
|
41
|
Zhang C, Yuan X, Mao W, Yue L, Kong X, Gao Y, Luo L, Yin Z. Inhibition of cadmium-induced apoptosis by glutathione S-transferase P1 via mitogen-activated protein kinases and mitochondrial pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2010; 30:202-208. [PMID: 21787653 DOI: 10.1016/j.etap.2010.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 05/20/2010] [Accepted: 06/25/2010] [Indexed: 05/31/2023]
Abstract
Cadmium is a well-known toxic metal for the kidney. Glutathione S-transferase P1 (GSTP1) plays an important role in the detoxification and xenobiotics metabolism. Here, we investigated whether GSTP1 affected Cd(2+)-induced apoptotic cell death in human embryonic kidney cell line (HEK) 293 cells. We showed that in HEK293 cells, silencing of GSTP1 expression through RNA interference reinforced the loss in cell viability induced by Cd(2+). Overexpression of GSTP1 inhibited loss of mitochondrial membrane potential, prevented cytochrome c release from mitochondria and caspase-3 activation, inhibited mitogen-activated protein kinases (MAPKs) including ERK, JNK and p38, and suppressed apoptosis induced by Cd(2+). The oligonucleosomal DNA fragmentation assay also demonstrated that overexpression of GSTP1 by adenovirus infection prevented Cd(2+)-induced apoptosis in primary renal tubule cells. Our data suggest that GSTP1 was an endogenous inhibitor of Cd(2+)-induced apoptosis.
Collapse
Affiliation(s)
- Chao Zhang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Dimethyl Sulfoxide Prevents 7β-Hydroxycholesterol-Induced Apoptosis by Preserving Lysosomes and Mitochondria. J Cardiovasc Pharmacol 2010; 56:263-7. [DOI: 10.1097/fjc.0b013e3181eb3063] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Chaudhary P, Sharma R, Sharma A, Vatsyayan R, Yadav S, Singhal SS, Rauniyar N, Prokai L, Awasthi S, Awasthi YC. Mechanisms of 4-hydroxy-2-nonenal induced pro- and anti-apoptotic signaling. Biochemistry 2010; 49:6263-75. [PMID: 20565132 DOI: 10.1021/bi100517x] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In recent years, 4-hydroxy-2-nonenal (4-HNE) has emerged as an important second messenger in cell cycle signaling. Here, we demonstrate that 4-HNE induces signaling for apoptosis via both the Fas-mediated extrinsic and the p53-mediated intrinsic pathways in HepG2 cells. 4-HNE induces a Fas-mediated DISC independent apoptosis pathway by activating ASK1, JNK, and caspase-3. Parallel treatment of 4-HNE to HepG2 cells also induces apoptosis by the p53 pathway through activation of Bax, p21, JNK, and caspase-3. Exposure of HepG2 cells to 4-HNE leads to the activation of both Fas and Daxx, promotes the export of Daxx from the nucleus to cytoplasm, and facilitates Fas-Daxx binding. Depletion of Daxx by siRNA results in the potentiation of apoptosis, indicating that Fas-Daxx binding in fact is inhibitory to Fas-mediated apoptosis in cells. 4-HNE-induced translocation of Daxx is also accompanied by the activation and nuclear accumulation of HSF1 and up-regulation of heat shock protein Hsp70. All these effects of 4-HNE in cells can be attenuated by ectopic expression of hGSTA4-4, the isozyme of glutathione S-transferase with high activity for 4-HNE. Through immunoprecipitation and liquid chromatography-tandem mass spectrometry, we have demonstrated the covalent binding of 4-HNE to Daxx. We also demonstrate that 4-HNE modification induces phosphorylation of Daxx at Ser668 and Ser671 to facilitate its cytoplasmic export. These results indicate that while 4-HNE exhibits toxicity through several mechanisms, in parallel it evokes signaling for defense mechanisms to self-regulate its toxicity and can simultaneously affect multiple signaling pathways through its interactions with membrane receptors and transcription factors/repressors.
Collapse
Affiliation(s)
- Pankaj Chaudhary
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Marí M, Colell A, Morales A, von Montfort C, Garcia-Ruiz C, Fernández-Checa JC. Redox control of liver function in health and disease. Antioxid Redox Signal 2010; 12:1295-331. [PMID: 19803748 PMCID: PMC2864660 DOI: 10.1089/ars.2009.2634] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reactive oxygen species (ROS), a heterogeneous population of biologically active intermediates, are generated as by-products of the aerobic metabolism and exhibit a dual role in biology. When produced in controlled conditions and in limited quantities, ROS may function as signaling intermediates, contributing to critical cellular functions such as proliferation, differentiation, and cell survival. However, ROS overgeneration and, particularly, the formation of specific reactive species, inflicts cell death and tissue damage by targeting vital cellular components such as DNA, lipids, and proteins, thus arising as key players in disease pathogenesis. Given the predominant role of hepatocytes in biotransformation and metabolism of xenobiotics, ROS production constitutes an important burden in liver physiology and pathophysiology and hence in the progression of liver diseases. Despite the recognized role of ROS in disease pathogenesis, the efficacy of antioxidants as therapeutics has been limited. A better understanding of the mechanisms, nature, and location of ROS generation, as well as the optimization of cellular defense strategies, may pave the way for a brighter future for antioxidants and ROS scavengers in the therapy of liver diseases.
Collapse
Affiliation(s)
- Montserrat Marí
- Liver Unit, Hospital Clinic, IDIBAPS-CIBEK, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Fabrini R, De Luca A, Stella L, Mei G, Orioni B, Ciccone S, Federici G, Lo Bello M, Ricci G. Monomer-dimer equilibrium in glutathione transferases: a critical re-examination. Biochemistry 2009; 48:10473-82. [PMID: 19795889 DOI: 10.1021/bi901238t] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glutathione transferases (GSTs) are dimeric enzymes involved in cell detoxification versus many endogenous toxic compounds and xenobiotics. In addition, single monomers of GSTs appear to be involved in particular protein-protein interactions as in the case of the pi class GST that regulates the apoptotic process by means of a GST-c-Jun N-terminal kinase complex. Thus, the dimer-monomer transition of GSTs may have important physiological relevance, but many studies reached contrasting conclusions both about the modality and extension of this event and about the catalytic competence of a single subunit. This paper re-examines the monomer-dimer question in light of novel experiments and old observations. Recent papers claimed the existence of a predominant monomeric and active species among pi, alpha, and mu class GSTs at 20-40 nM dilution levels, reporting dissociation constants (K(d)) for dimeric GST of 5.1, 0.34, and 0.16 microM, respectively. However, we demonstrate here that only traces of monomers could be found at these concentrations since all these enzymes display K(d) values of <<1 nM, values thousands of times lower than those reported previously. Time-resolved and steady-state fluorescence anisotropy experiments, two-photon fluorescence correlation spectroscopy, kinetic studies, and docking simulations have been used to reach such conclusions. Our results also indicate that there is no clear evidence of the existence of a fully active monomer. Conversely, many data strongly support the idea that the monomeric form is scarcely active or fully inactive.
Collapse
Affiliation(s)
- Raffaele Fabrini
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Taki K, Shimozono R, Kusano H, Suzuki N, Shinjo K, Eda H. Apoptosis signal-regulating kinase 1 is crucial for oxidative stress-induced but not for osmotic stress-induced hepatocyte cell death. Life Sci 2008; 83:859-64. [DOI: 10.1016/j.lfs.2008.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 09/23/2008] [Accepted: 10/03/2008] [Indexed: 01/01/2023]
|
47
|
Case J, Ingram DA, Haneline LS. Oxidative stress impairs endothelial progenitor cell function. Antioxid Redox Signal 2008; 10:1895-907. [PMID: 18627346 PMCID: PMC2575029 DOI: 10.1089/ars.2008.2118] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Circulating endothelial progenitor cells (EPCs) in adult human peripheral blood were identified in 1997. Since their original identification, EPCs have been extensively studied as biomarkers to assess the risk of cardiovascular disease in human subjects and as a potential cell therapeutic for vascular regeneration. EPCs are exposed to oxidative stress during vascular injury as residents of blood vessel walls or as circulating cells homing to sites of neovascularization. Given the links between oxidative injury, endothelial cell dysfunction, and vascular disease, recent investigation has focused on the responses of EPCs to oxidant stress and the molecular mechanisms that control redox regulation in these specialized cells. In this review, we discuss the various cell and flow-cytometric techniques used to define and isolate EPCs from circulating blood and the current human and mouse genetic data, which offer insights into redox control in EPC biology and angiogenesis. Finally, we review how EPC responses to oxidant stress may be a critical determinant in maintaining the integrity and function of the cardiovascular system and how perturbations of redox control in EPCs may lead to various human diseases.
Collapse
Affiliation(s)
- Jamie Case
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
48
|
Luo L, Wang Y, Feng Q, Zhang H, Xue B, Shen J, Ye Y, Han X, Ma H, Xu J, Chen D, Yin Z. Recombinant protein glutathione S-transferases P1 attenuates inflammation in mice. Mol Immunol 2008; 46:848-57. [PMID: 18962899 DOI: 10.1016/j.molimm.2008.09.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/10/2008] [Accepted: 09/07/2008] [Indexed: 10/21/2022]
Abstract
We have reported that intracellular glutathione S-transferases P1 (GSTP1) suppresses LPS (lipopolysaccharide)-induced excessive production of pro-inflammatory factors by inhibiting LPS-stimulated MAPKs (mitogen-activated protein kinases) as well as NF-kappaB activation. But under pathogenic circumstances, physiologic levels of GSTP1 are insufficient to stem pro-inflammatory signaling. Here we show that LPS-induced up-regulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in RAW246.7 cells is significantly reduced by incubating cells with recombinant GSTP1 protein. In vivo study demonstrates that treatment of mice (i.p.) with recombinant GSTP1 protein effectively suppresses the devastating effects of acute inflammation, which includes reduction of mortality rate of endotoxic shock, alleviation of LPS-induced acute lung injury and abrogation of thioglycolate (TG)-induced peritoneal deposition of leukocytes and polymorphonuclear cells (PMNs). Meanwhile, GSTP1 prevented LPS-induced TNF-alpha, IL-1beta, MCP-1 and NO production. Further investigation by using confocal microscopy and flow cytometry shows that recombinant GSTP1 protein can be delivered into RAW246.7 cells, mouse peritoneal macrophages and HEK 293 cells suggesting that extracellular GSTP1 protein could be transported across plasma membrane and act as a cytosolic protein. In conclusion our research demonstrates a new finding that increasing cellular GSTP1 level by supplement of recombinant GSTP1 effectively suppresses the devastating effects of acute inflammation.
Collapse
Affiliation(s)
- Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fujikawa Y, Urano Y, Komatsu T, Hanaoka K, Kojima H, Terai T, Inoue H, Nagano T. Design and Synthesis of Highly Sensitive Fluorogenic Substrates for Glutathione S-Transferase and Application for Activity Imaging in Living Cells. J Am Chem Soc 2008; 130:14533-43. [DOI: 10.1021/ja802423n] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Yuuta Fujikawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hirotatsu Kojima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takuya Terai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hideshi Inoue
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tetsuo Nagano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan, and School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
50
|
Kim SH, Sharma RP. Mercury Alters Endotoxin-Induced Inflammatory Cytokine Expression in Liver: Differential Roles of P38 and Extracellular Signal-Regulated Mitogen-Activated Protein Kinases. Immunopharmacol Immunotoxicol 2008; 27:123-35. [PMID: 15803865 DOI: 10.1081/iph-51757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Mercury is a widespread metal in the environment and consequently large populations are currently exposed to low levels of mercury. Endotoxin, a component of the Gram-negative bacteria, promotes inflammatory responses. We recently reported that mercury modulates the production of nitric oxide and various inflammatory cytokines induced by endotoxin in a macrophage cell line (Nitric Oxide 2002, 7:67). The present study was designed to determine the impact of mercury on endotoxin-induced inflammatory cytokine expression and corresponding signal transduction in mouse liver. Male BALB/c mice were exposed continuously to 0, 0.3, 1.5, 7.5, or 37.5 ppm of mercury in drinking water for 14 days and at the end of the treatment period lipopolysaccharide (LPS, 0.5 mg/kg) was injected intraperitoneally 2 hr prior to euthanasia. The doses of mercury and LPS did not cause hepatotoxicity as indicated by unaltered circulating alanine aminotransferase and aspartate aminotransferase levels. Mercury decreased liver glutathione (GSH) and with LPS additively decreased GSH. Mercury activated p38 mitogen-activated protein kinase (MAPK) and additively increased LPS-induced p38 MAPK phosphorylation. In contrast, mercury alone had no effect on activation of extracellular signal-regulated kinase (ERK) but inhibited LPS-induced ERK activation. Mercury increased the expression of tumor necrosis factor alpha (TNFalpha) and further potentiated LPS-induced TNFalpha expression. Mercury did not affect LPS-induced interleukin (IL)-1beta expression but decreased LPS-induced IL-6 expression. Results indicated that low levels of mercury augment LPS-induced TNFalpha expression by altering GSH and p38 MAPK. Mercury modulates LPS-induced p38 and ERK activation and downstream TNFalpha and IL-6 expression in mouse liver.
Collapse
Affiliation(s)
- Sang Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | | |
Collapse
|