1
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [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: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
2
|
Yang J, Chen X. SIRT6 attenuates LPS-induced inflammation and apoptosis of lung epithelial cells in acute lung injury through ACE2/STAT3/PIM1 signaling. Immun Inflamm Dis 2023; 11:e809. [PMID: 36988243 PMCID: PMC10022422 DOI: 10.1002/iid3.809] [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: 11/09/2022] [Revised: 01/10/2023] [Accepted: 02/18/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a severe and fatal respiratory disease. SIRT6 exerts pivotal activities in the process of lung diseases, but whether SIRT6 impacts ALI has not been covered. METHODS Lentivirus recombinant expressing vector SIRT6 gene (Lent-SIRT6) was constructed in mice, and there were control, lipopolysaccharide (LPS), LPS + Vehicle, and LPS + Lent SIRT6 groups. RT-qPCR and western blot detected SIRT6 expression in lung tissues. HE staining observed pathological alternations in lung tissues. Wet-to-dry ratio of the lungs was then measured. The cell count of bronchoalveolar lavage fluid (BALF) was evaluated. Serum inflammation was examined with enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and western blot were to measure apoptosis. Western blot tested the expression of ACE2/STAT3/PIM1 signaling-associated factors. At the cellular level, LPS was used to induce lung epithelial cells BEAS-2B to establish cell injury models. SIRT6 was overexpressed and ACE2 expression was inhibited by cell transfection, and the mechanism of SIRT6 in LPS-induced lung injury model was further explored by Cell Counting Kit-8 (CCK-8), western blot, quantitative reverse-transcription polymerase chain reaction, TUNEL, and other techniques. RESULTS The results of animal experiments showed that SIRT6 overexpression could reduce LPS-induced lung pathological injury, pulmonary edema, and BALF cell ratio and attenuate LPS-induced inflammatory response and cell apoptosis. In the above process, ACE2, STAT3, p-STAT3, and PIM1 expression were affected. In cell experiments, SIRT6 expression was reduced in LPS-induced BEAS-2B cells. Inhibition of ACE2 expression could reverse the inhibitory effect of SIRT6 overexpression on ACE2/STAT3/PIM1 pathway, and cellular inflammatory response and apoptosis. CONCLUSION SIRT6 eased LPS-evoked inflammation and apoptosis of lung epithelial cells in ALI through ACE2/STAT3/PIM1 signaling.
Collapse
Affiliation(s)
- Juan Yang
- Department of Pediatric, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xing Chen
- Department of Pediatric, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| |
Collapse
|
3
|
Gomez HM, Pillar AL, Brown AC, Kim RY, Ali MK, Essilfie AT, Vanders RL, Frazer DM, Anderson GJ, Hansbro PM, Collison AM, Jensen ME, Murphy VE, Johnstone DM, Reid D, Milward EA, Donovan C, Horvat JC. Investigating the Links between Lower Iron Status in Pregnancy and Respiratory Disease in Offspring Using Murine Models. Nutrients 2021; 13:nu13124461. [PMID: 34960012 PMCID: PMC8708709 DOI: 10.3390/nu13124461] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Maternal iron deficiency occurs in 40-50% of all pregnancies and is associated with an increased risk of respiratory disease and asthma in children. We used murine models to examine the effects of lower iron status during pregnancy on lung function, inflammation and structure, as well as its contribution to increased severity of asthma in the offspring. A low iron diet during pregnancy impairs lung function, increases airway inflammation, and alters lung structure in the absence and presence of experimental asthma. A low iron diet during pregnancy further increases these major disease features in offspring with experimental asthma. Importantly, a low iron diet increases neutrophilic inflammation, which is indicative of more severe disease, in asthma. Together, our data demonstrate that lower dietary iron and systemic deficiency during pregnancy can lead to physiological, immunological and anatomical changes in the lungs and airways of offspring that predispose to greater susceptibility to respiratory disease. These findings suggest that correcting iron deficiency in pregnancy using iron supplements may play an important role in preventing or reducing the severity of respiratory disease in offspring. They also highlight the utility of experimental models for understanding how iron status in pregnancy affects disease outcomes in offspring and provide a means for testing the efficacy of different iron supplements for preventing disease.
Collapse
Affiliation(s)
- Henry M. Gomez
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Amber L. Pillar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Alexandra C. Brown
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Richard Y. Kim
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Md Khadem Ali
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Ama-Tawiah Essilfie
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
| | - Rebecca L. Vanders
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - David M. Frazer
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Gregory J. Anderson
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Philip M. Hansbro
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Adam M. Collison
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Megan E. Jensen
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Vanessa E. Murphy
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Daniel M. Johnstone
- School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia;
| | - David Reid
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
| | - Elizabeth A. Milward
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Chantal Donovan
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jay C. Horvat
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Correspondence: ; Tel.: +612-4042-0220
| |
Collapse
|
4
|
Tirunavalli SK, Gourishetti K, Kotipalli RSS, Kuncha M, Kathirvel M, Kaur R, Jerald MK, Sistla R, Andugulapati SB. Dehydrozingerone ameliorates Lipopolysaccharide induced acute respiratory distress syndrome by inhibiting cytokine storm, oxidative stress via modulating the MAPK/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153729. [PMID: 34517257 PMCID: PMC8390101 DOI: 10.1016/j.phymed.2021.153729] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND Inflammation-mediated lung injury is a major cause of health problems in many countries and has been the leading cause of morbidity/mortality in intensive care units. In the current COVID-19 pandemic, the majority of the patients experienced serious pneumonia resulting from inflammation (Acute respiratory distress syndrome/ARDS). Pathogenic infections cause cytokine release syndrome (CRS) by hyperactivation of immune cells, which in turn release excessive cytokines causing ARDS. Currently, there are no standard therapies for viral, bacterial or pathogen-mediated CRS. PURPOSE This study aimed to investigate and validate the protective effects of Dehydrozingerone (DHZ) against LPS induced lung cell injury by in-vitro and in-vivo models and to gain insights into the molecular mechanisms that mediate these therapeutic effects. METHODS The therapeutic activity of DHZ was determined in in-vitro models by pre-treating the cells with DHZ and exposed to LPS to stimulate the inflammatory cascade of events. We analysed the effect of DHZ on LPS induced inflammatory cytokines, chemokines and cell damage markers expression/levels using various cell lines. We performed gene expression, ELISA, and western blot analysis to elucidate the effect of DHZ on inflammation and its modulation of MAPK and NF-κB pathways. Further, the prophylactic and therapeutic effect of DHZ was evaluated against the LPS induced ARDS model in rats. RESULTS DHZ significantly (p < 0.01) attenuated the LPS induced ROS, inflammatory cytokine, chemokine gene expression and protein release in macrophages. Similarly, DHZ treatment protected the lung epithelial and endothelial cells by mitigating the LPS induced inflammatory events in a dose-dependent manner. In vivo analysis showed that DHZ treatment significantly (p < 0.001) mitigated the LPS induced ARDS pathophysiology of increase in the inflammatory cells in BALF, inflammatory cytokine and chemokines in lung tissues. LPS stimulated neutrophil-mediated events, apoptosis, alveolar wall thickening and alveolar inflammation were profoundly reduced by DHZ treatment in a rat model. CONCLUSION This study demonstrates for the first time that DHZ has the potential to ameliorate LPS induced ARDS by inhibiting cytokine storm and oxidative through modulating the MAPK and NF-κB pathways. This data provides pre-clinical support to develop DHZ as a potential therapeutic agent against ARDS.
Collapse
Affiliation(s)
- Satya Krishna Tirunavalli
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Karthik Gourishetti
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Madusudhana Kuncha
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | | | - Rajwinder Kaur
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | - Mahesh Kumar Jerald
- CSIR - Centre for Cellular & Molecular Biology (CCMB), Hyderabad 500 007, India
| | - Ramakrishna Sistla
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Sai Balaji Andugulapati
- CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Centre for Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.
| |
Collapse
|
5
|
Xi Y, Li Y, Xu P, Li S, Liu Z, Tung HC, Cai X, Wang J, Huang H, Wang M, Xu M, Ren S, Li S, Zhang M, Lee YJ, Huang L, Yang D, He J, Huang Z, Xie W. The anti-fibrotic drug pirfenidone inhibits liver fibrosis by targeting the small oxidoreductase glutaredoxin-1. SCIENCE ADVANCES 2021; 7:eabg9241. [PMID: 34516906 PMCID: PMC8442864 DOI: 10.1126/sciadv.abg9241] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/08/2021] [Indexed: 02/05/2023]
Abstract
Activation of the hepatic stellate cells (HSCs) is a key pathogenic event in liver fibrosis. Protein S-glutathionylation (PSSG) of cysteine residues is a distinct form of oxidative response that modifies protein structures and functions. Glutaredoxin-1 (GLRX) reverses PSSG by liberating glutathione (GSH). In this study, we showed that pirfenidone (PFD), an anti-lung fibrosis drug, inhibited HSC activation and liver fibrosis in a GLRX-dependent manner. Glrx depletion exacerbated liver fibrosis, and decreased GLRX and increased PSSG were observed in fibrotic mouse and human livers. In contrast, overexpression of GLRX inhibited PSSG and liver fibrosis. Mechanistically, the inhibition of HSC activation by GLRX may have been accounted for by deglutathionylation of Smad3, which inhibits Smad3 phosphorylation, leading to the suppression of fibrogenic gene expression. Our results have established GLRX as the therapeutic target of PFD and uncovered an important role of PSSG in liver fibrosis. GLRX/PSSG can be both a biomarker and a therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Yue Xi
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanping Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sihan Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zhengsheng Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hung-chun Tung
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xinran Cai
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jingyuan Wang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Haozhe Huang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Song Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Min Zhang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yong J. Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Da Yang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
6
|
Seyrek K, Lavrik IN. Modulation of CD95-mediated signaling by post-translational modifications: towards understanding CD95 signaling networks. Apoptosis 2020; 24:385-394. [PMID: 31069559 DOI: 10.1007/s10495-019-01540-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CD95 is a member of the death receptor family and is well-known to promote apoptosis. However, accumulating evidence indicates that in some context CD95 has not only the potential to induce apoptosis but also can trigger non-apoptotic signal leading to cell survival, proliferation, cancer growth and metastasis. Despite extensive investigations focused on alterations in the expression level of CD95 and associated signal molecules, very few studies, however, have investigated the effects of post-translational modifications such as glycosylation, phosphorylation, palmitoylation, nitrosylation and glutathionylation on CD95 function. Post-translational modifications of CD95 in mammalian systems are likely to play a more prominent role than anticipated in CD95 induced cell death. In this review we will focus on the alterations in CD95-mediated signaling caused by post-translational modifications of CD95.
Collapse
Affiliation(s)
- Kamil Seyrek
- Translational Inflammation Research, Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Inna N Lavrik
- Translational Inflammation Research, Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany.
| |
Collapse
|
7
|
Zheng X, Li Q, Tian H, Li H, Lv Y, Wang Y, He L, Huo Y, Hao Z. HIP/PAP protects against bleomycin-induced lung injury and inflammation and subsequent fibrosis in mice. J Cell Mol Med 2020; 24:6804-6821. [PMID: 32352211 PMCID: PMC7299702 DOI: 10.1111/jcmm.15334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/10/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
Hepatocarcinoma‐intestine‐pancreas/pancreatitis‐associated protein (HIP/PAP), a C‐type lectin, exerts anti‐oxidative, anti‐inflammatory, bactericidal, anti‐apoptotic, and mitogenic functions in several cell types and tissues. In this study, we explored the role of HIP/PAP in pulmonary fibrosis (PF). Expression of HIP/PAP and its murine counterpart, Reg3B, was markedly increased in fibrotic human and mouse lung tissues. Adenovirus‐mediated HIP/PAP expression markedly alleviated bleomycin (BLM)‐induced lung injury, inflammation, and fibrosis in mice. Adenovirus‐mediated HIP/PAP expression alleviated oxidative injury and lessened the decrease in pulmonary superoxide dismutase (SOD) activity in BLM‐treated mice, increased pulmonary SOD expression in normal mice, and HIP/PAP upregulated SOD expression in cultured human alveolar epithelial cells (A549) and human lung fibroblasts (HLF‐1). Moreover, in vitro experiments showed that HIP/PAP suppressed the growth of HLF‐1 and ameliorated the H2O2‐induced apoptosis of human alveolar epithelial cells (A549 and HPAEpiC) and human pulmonary microvascular endothelial cells (HPMVEC). In HLF‐1, A549, HPAEpiC, and HPMVEC cells, HIP/PAP did not affect the basal levels, but alleviated the TGF‐β1‐induced down‐regulation of the epithelial/endothelial markers E‐cadherin and vE‐cadherin and the over‐expression of mesenchymal markers, such as α‐SMA and vimentin. In conclusion, HIP/PAP was found to serve as a potent protective factor in lung injury, inflammation, and fibrosis by attenuating oxidative injury, promoting the regeneration of alveolar epithelial cells, and antagonizing the pro‐fibrotic actions of the TGF‐β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xiaoyan Zheng
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qian Li
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong Tian
- Research Center of Reproductive Medicine, Medical School of Xi'an Jiaotong University, Xi'an, China
| | - Hanchao Li
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yifei Lv
- Department of Gastroenterology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanhua Wang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lan He
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongwei Huo
- Research Center of Reproductive Medicine, Medical School of Xi'an Jiaotong University, Xi'an, China
| | - Zhiming Hao
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
9
|
He N, Bai S, Huang Y, Xing Y, Chen L, Yu F, Lv C. Evaluation of Glutathione S-Transferase Inhibition Effects on Idiopathic Pulmonary Fibrosis Therapy with a Near-Infrared Fluorescent Probe in Cell and Mice Models. Anal Chem 2019; 91:5424-5432. [PMID: 30869868 DOI: 10.1021/acs.analchem.9b00713] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung-limited and progressive fibrotic disease. The early diagnosis and therapies of IPF are still full of clinical challenges. Glutathione S-transferase (GSTs) plays significant roles in promoting the formation of pulmonary fibrosis. Herein, we report a fluorescent probe (Cy-GST) for the detection of GSTs concentration fluctuations in cells and in mice models. The probe can selectively and sensitively respond to GSTs with an "off-on" type fluorescence switch. Our results demonstrated that the level of intracellular GSTs increase in the pulmonary fibrosis cells and mice models. And the IPF patients hold high levels of GSTs concentrations. Thus, GSTs are likely to play important roles in pulmonary fibrosis. The inhibitor of GSTs TLK117 can reduce the severity of pulmonary fibrosis. The synergistic treatment of TLK117 and pirfenidone have better therapeutic effects than only using pirfenidone in pulmonary fibrosis mice models. The level of GSTs in IPF may be a new potential marker for IPF diagnosis. And the inhibition of GSTs may be a new therapeutic strategy for IPF treatment.
Collapse
Affiliation(s)
- Na He
- Department of Respiratory Medicine , Binzhou Medical University Hospital , Binzhou 256603 , China.,Medicine Research Center, Institute of Molecular Medicine , Binzhou Medical University , Yantai 264003 , China
| | - Song Bai
- Department of Respiratory Medicine , Binzhou Medical University Hospital , Binzhou 256603 , China.,Medicine Research Center, Institute of Molecular Medicine , Binzhou Medical University , Yantai 264003 , China
| | - Yan Huang
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research , Chinese Academy of Sciences , Yantai 264003 , China
| | - Yanlong Xing
- Institute of Functional Materials and Molecular Imaging, College of Clinical Medicine, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma , Hainan Medical University , Haikou 571199 , China
| | - Lingxin Chen
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research , Chinese Academy of Sciences , Yantai 264003 , China
| | - Fabiao Yu
- Institute of Functional Materials and Molecular Imaging, College of Clinical Medicine, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma , Hainan Medical University , Haikou 571199 , China
| | - Changjun Lv
- Department of Respiratory Medicine , Binzhou Medical University Hospital , Binzhou 256603 , China.,Medicine Research Center, Institute of Molecular Medicine , Binzhou Medical University , Yantai 264003 , China
| |
Collapse
|
10
|
Anathy V, Lahue KG, Chapman DG, Chia SB, Casey DT, Aboushousha R, van der Velden JLJ, Elko E, Hoffman SM, McMillan DH, Jones JT, Nolin JD, Abdalla S, Schneider R, Seward DJ, Roberson EC, Liptak MD, Cousins ME, Butnor KJ, Taatjes DJ, Budd RC, Irvin CG, Ho YS, Hakem R, Brown KK, Matsui R, Bachschmid MM, Gomez JL, Kaminski N, van der Vliet A, Janssen-Heininger YMW. Reducing protein oxidation reverses lung fibrosis. Nat Med 2018; 24:1128-1135. [PMID: 29988126 PMCID: PMC6204256 DOI: 10.1038/s41591-018-0090-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis is characterized by excessive deposition of collagen in the lung, leading to chronically impaired gas exchange and death1-3. Oxidative stress is believed to be critical in this disease pathogenesis4-6, although the exact mechanisms remain enigmatic. Protein S-glutathionylation (PSSG) is a post-translational modification of proteins that can be reversed by glutaredoxin-1 (GLRX)7. It remains unknown whether GLRX and PSSG play a role in lung fibrosis. Here, we explored the impact of GLRX and PSSG status on the pathogenesis of pulmonary fibrosis, using lung tissues from subjects with idiopathic pulmonary fibrosis, transgenic mouse models and direct administration of recombinant Glrx to airways of mice with existing fibrosis. We demonstrate that GLRX enzymatic activity was strongly decreased in fibrotic lungs, in accordance with increases in PSSG. Mice lacking Glrx were far more susceptible to bleomycin- or adenovirus encoding active transforming growth factor beta-1 (AdTGFB1)-induced pulmonary fibrosis, whereas transgenic overexpression of Glrx in the lung epithelium attenuated fibrosis. We furthermore show that endogenous GLRX was inactivated through an oxidative mechanism and that direct administration of the Glrx protein into airways augmented Glrx activity and reversed increases in collagen in mice with TGFB1- or bleomycin-induced fibrosis, even when administered to fibrotic, aged animals. Collectively, these findings suggest the therapeutic potential of exogenous GLRX in treating lung fibrosis.
Collapse
Affiliation(s)
- Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David G Chapman
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Dylan T Casey
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jane T Jones
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sarah Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Robert Schneider
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David J Seward
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | | - Matthew D Liptak
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Morgan E Cousins
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Kelly J Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Ralph C Budd
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Charles G Irvin
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Razq Hakem
- Department of Medical Biophysics and Immunology, University of Toronto, and the Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Kevin K Brown
- Department of Medicine, Pulmonary, Critical Care and Sleep Section, National Jewish Health and the University of Colorado, Denver, CO, USA
| | - Reiko Matsui
- Department of Medicine, Boston University, Boston, MA, USA
| | | | - Jose L Gomez
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
11
|
Propofol inhibits LPS-induced apoptosis in lung epithelial cell line, BEAS-2B. Biomed Pharmacother 2017; 87:180-187. [DOI: 10.1016/j.biopha.2016.12.074] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/11/2016] [Accepted: 12/19/2016] [Indexed: 11/23/2022] Open
|
12
|
Identification and In Silico Analysis of Major Redox Modulated Proteins from Brassica juncea Seedlings Using 2D Redox SDS PAGE (2-Dimensional Diagonal Redox Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis). Protein J 2017; 36:64-76. [DOI: 10.1007/s10930-017-9698-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
13
|
Evidence for a role for the putative Drosophila hGRX1 orthologue in copper homeostasis. Biometals 2016; 29:705-13. [PMID: 27379771 DOI: 10.1007/s10534-016-9946-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 06/29/2016] [Indexed: 12/18/2022]
Abstract
Glutaredoxins are a family of small molecular weight proteins that have a central role in cellular redox regulation. Human GRX1 (hGRX1) has also been shown to play an integral role in copper homeostasis by regulating the redox activity of the metalated sites of copper chaperones such as ATOX1 and SOD1, and the copper efflux proteins ATP7A and ATP7B. To further elucidate the role of hGRX1 in copper homeostasis, we examined the impact of RNA interference-mediated knockdown of CG6852, a putative Drosophila orthologue of hGRX1. CG6852 shares ~41 % amino acid identity with hGRX1 and key functional domains including the metal-binding CXXC motif are conserved between the two proteins. Knockdown of CG6852 in the adult midline caused a thoracic cleft and reduced scutellum, phenotypes that were exacerbated by additional knockdown of copper uptake transporters Ctr1A and Ctr1B. Knockdown of CG6852 in the adult eye enhanced a copper-deficiency phenotype caused by Ctr1A knockdown while ubiquitous knockdown of CG6852 resulted a mild systemic copper deficiency. Therefore we conclude that CG6852 is a putative orthologue of hGRX1 and may play an important role in Drosophila copper homeostasis.
Collapse
|
14
|
McMillan DH, van der Velden JL, Lahue KG, Qian X, Schneider RW, Iberg MS, Nolin JD, Abdalla S, Casey DT, Tew KD, Townsend DM, Henderson CJ, Wolf CR, Butnor KJ, Taatjes DJ, Budd RC, Irvin CG, van der Vliet A, Flemer S, Anathy V, Janssen-Heininger YM. Attenuation of lung fibrosis in mice with a clinically relevant inhibitor of glutathione- S-transferase π. JCI Insight 2016; 1:85717. [PMID: 27358914 PMCID: PMC4922427 DOI: 10.1172/jci.insight.85717] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/04/2016] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating lung disease characterized by excessive collagen production and fibrogenesis. Apoptosis in lung epithelial cells is critical in IPF pathogenesis, as heightened loss of these cells promotes fibroblast activation and remodeling. Changes in glutathione redox status have been reported in IPF patients. S-glutathionylation, the conjugation of glutathione to reactive cysteines, is catalyzed in part by glutathione-S-transferase π (GSTP). To date, no published information exists linking GSTP and IPF to our knowledge. We hypothesized that GSTP mediates lung fibrogenesis in part through FAS S-glutathionylation, a critical event in epithelial cell apoptosis. Our results demonstrate that GSTP immunoreactivity is increased in the lungs of IPF patients, notably within type II epithelial cells. The FAS-GSTP interaction was also increased in IPF lungs. Bleomycin- and AdTGFβ-induced increases in collagen content, α-SMA, FAS S-glutathionylation, and total protein S-glutathionylation were strongly attenuated in Gstp-/- mice. Oropharyngeal administration of the GSTP inhibitor, TLK117, at a time when fibrosis was already apparent, attenuated bleomycin- and AdTGFβ-induced remodeling, α-SMA, caspase activation, FAS S-glutathionylation, and total protein S-glutathionylation. GSTP is an important driver of protein S-glutathionylation and lung fibrosis, and GSTP inhibition via the airways may be a novel therapeutic strategy for the treatment of IPF.
Collapse
Affiliation(s)
- David H. McMillan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Jos L.J. van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Karolyn G. Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Robert W. Schneider
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Martina S. Iberg
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - James D. Nolin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sarah Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Dylan T. Casey
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Kenneth D. Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Danyelle M. Townsend
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Colin J. Henderson
- Division of Cancer Research, University of Dundee, Dundee, United Kingdom
| | - C. Roland Wolf
- Division of Cancer Research, University of Dundee, Dundee, United Kingdom
| | - Kelly J. Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Douglas J. Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | | | | | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Stevenson Flemer
- Department of Chemistry, University of Vermont, Burlington, Vermont, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | | |
Collapse
|
15
|
Inhibition of endotoxin-induced airway epithelial cell injury by a novel family of pyrrol derivates. J Transl Med 2016; 96:632-40. [PMID: 26999659 DOI: 10.1038/labinvest.2016.46] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/18/2016] [Accepted: 02/02/2016] [Indexed: 01/14/2023] Open
Abstract
Inflammation and apoptosis are crucial mechanisms for the development of the acute respiratory distress syndrome (ARDS). Currently, there is no specific pharmacological therapy for ARDS. We have evaluated the ability of a new family of 1,2,3,5-tetrasubstituted pyrrol compounds for attenuating lipopolysaccharide (LPS)-induced inflammation and apoptosis in an in vitro LPS-induced airway epithelial cell injury model based on the first steps of the development of sepsis-induced ARDS. Human alveolar A549 and human bronchial BEAS-2B cells were exposed to LPS, either alone or in combination with the pyrrol derivatives. Rhein and emodin, two representative compounds with proven activity against the effects of LPS, were used as reference compounds. The pyrrol compound that was termed DTA0118 had the strongest inhibitory activity and was selected as the lead compound to further explore its properties. Exposure to LPS caused an intense inflammatory response and apoptosis in both A549 and BEAS-2B cells. DTA0118 treatment downregulated Toll-like receptor-4 expression and upregulated nuclear factor-κB inhibitor-α expression in cells exposed to LPS. These anti-inflammatory effects were accompanied by a significantly lower secretion of interleukin-6 (IL-6), IL-8, and IL-1β. The observed antiapoptotic effect of DTA0118 was associated with the upregulation of antiapoptotic Bcl-2 and downregulation of proapoptotic Bax and active caspase-3 protein levels. Our findings demonstrate the potent anti-inflammatory and antiapoptotic properties of the pyrrol DTA0118 compound and suggest that it could be considered as a potential drug therapy for the acute phase of sepsis and septic ARDS. Further investigations are needed to examine and validate these mechanisms and effects in a clinically relevant animal model of sepsis and sepsis-induced ARDS.
Collapse
|
16
|
Jaramillo MC, Briehl MM, Batinic-Haberle I, Tome ME. Manganese (III) meso-tetrakis N-ethylpyridinium-2-yl porphyrin acts as a pro-oxidant to inhibit electron transport chain proteins, modulate bioenergetics, and enhance the response to chemotherapy in lymphoma cells. Free Radic Biol Med 2015; 83:89-100. [PMID: 25725417 PMCID: PMC4441837 DOI: 10.1016/j.freeradbiomed.2015.01.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/22/2015] [Accepted: 01/29/2015] [Indexed: 11/23/2022]
Abstract
The manganese porphyrin, manganese (III) meso-tetrakis N-ethylpyridinium-2-yl porphyrin (MnTE-2-PyP(5+)), acts as a pro-oxidant in the presence of intracellular H2O2. Mitochondria are the most prominent source of intracellular ROS and important regulators of the intrinsic apoptotic pathway. Due to the increased oxidants near and within the mitochondria, we hypothesized that the mitochondria are a target of the pro-oxidative activity of MnTE-2-PyP(5+) and that we could exploit this effect to enhance the chemotherapeutic response in lymphoma. In this study, we demonstrate that MnTE-2-PyP(5+) modulates the mitochondrial redox environment and sensitizes lymphoma cells to antilymphoma chemotherapeutics. MnTE-2-PyP(5+) increased dexamethasone-induced mitochondrial ROS and oxidation of the mitochondrial glutathione pool in lymphoma cells. The combination treatment induced glutathionylation of Complexes I, III, and IV in the electron transport chain, and decreased the activity of Complexes I and III, but not the activity of Complex IV. Treatment with the porphyrin and dexamethasone also decreased cellular ATP levels. Rho(0) malignant T-cells with impaired mitochondrial electron transport chain function were less sensitive to the combination treatment than wild-type cells. These findings suggest that mitochondria are important for the porphyrin's ability to enhance cell death. MnTE-2-PyP(5+) also augmented the effects of 2-deoxy-D-glucose (2DG), an antiglycolytic agent. In combination with 2DG, MnTE-2-PyP(5+) increased protein glutathionylation, decreased ATP levels more than 2DG treatment alone, and enhanced 2DG-induced cell death in primary B-ALL cells. MnTE-2-PyP(5+) did not enhance dexamethasone- or 2DG-induced cell death in normal cells. Our findings suggest that MnTE-2-PyP(5+) has potential as an adjuvant for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Melba C Jaramillo
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA
| | - Margaret M Briehl
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Margaret E Tome
- Medical Pharmacology, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
17
|
|
18
|
Glutathione and redox signaling in substance abuse. Biomed Pharmacother 2014; 68:799-807. [PMID: 25027386 DOI: 10.1016/j.biopha.2014.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 06/12/2014] [Indexed: 01/04/2023] Open
Abstract
Throughout the last couple decades, the cause and consequences of substance abuse has expanded to identify the underlying neurobiological signaling mechanisms associated with addictive behavior. Chronic use of drugs, such as cocaine, methamphetamine and alcohol leads to the formation of oxidative or nitrosative stress (ROS/RNS) and changes in glutathione and redox homeostasis. Of importance, redox-sensitive post-translational modifications on cysteine residues, such as S-glutathionylation and S-nitrosylation could impact on the structure and function of addiction related signaling proteins. In this commentary, we evaluate the role of glutathione and redox signaling in cocaine-, methamphetamine- and alcohol addiction and conclude by discussing the possibility of targeting redox pathways for the therapeutic intervention of these substance abuse disorders.
Collapse
|
19
|
|
20
|
Abstract
SIGNIFICANCE Glutaredoxins (Grxs) are small oxidoreductases of the thioredoxin family of proteins regulating the thiol redox state of several proteins. Thereby, Grxs are key elements in redox signaling. RECENT ADVANCES Redox signaling via protein thiols depends on reversible oxidative modifications induced mainly by reactive oxygen/nitrogen species and glutathione (GSH) in form of its oxidized disulfide or S-nitroso-glutathione. Grxs contribute to redox signaling by the catalysis of glutathionylation, de-glutathionylation, as well as reduction of disulfide bridges via two distinct enzymatic mechanisms. The dithiol mechanism utilizes both active site cysteines to reduce disulfides, whereas the monothiol mechanism utilizes only the N-terminal active site cysteine for the reduction of GSH mixed disulfides. The sphere of action of Grxs continues to grow with the recent identification of novel targets. CRITICAL ISSUES Because of limited methodological tools, the identification of new substrates for oxidoreductases in general is one of the biggest challenges in this research area. FUTURE DIRECTIONS With this review, we provide a condensed summary of the current knowledge of thiol/disulfide exchange reactions catalyzed by Grxs regarding the mechanistic, structural, and functional aspects. The latter will be of high importance for future research directions, gaining novel insights into redox signaling in general, and the role of Grxs in particular.
Collapse
Affiliation(s)
- Christopher Horst Lillig
- Institut für Biochemie und Molekularbiologie, Universitätsmedizin Greifswald, Ernst Moritz Arndt-Universität Greifswald, Greifswald, Germany
| | | |
Collapse
|
21
|
Wei W, Lampe L, Park S, Vangara BS, Waldo GS, Cabantous S, Subaran SS, Yang D, Lakatta EG, Lin L. Disulfide bonds within the C2 domain of RAGE play key roles in its dimerization and biogenesis. PLoS One 2012; 7:e50736. [PMID: 23284645 PMCID: PMC3524233 DOI: 10.1371/journal.pone.0050736] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 10/22/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The receptor for advanced glycation end products (RAGE) on the cell surface transmits inflammatory signals. A member of the immunoglobulin superfamily, RAGE possesses the V, C1, and C2 ectodomains that collectively constitute the receptor's extracellular structure. However, the molecular mechanism of RAGE biogenesis remains unclear, impeding efforts to control RAGE signaling through cellular regulation. METHODOLOGY AND RESULT: We used co-immunoprecipitation and crossing-linking to study RAGE oligomerization and found that RAGE forms dimer-based oligomers. Via non-reducing SDS-polyacrylamide gel electrophoresis and mutagenesis, we found that cysteines 259 and 301 within the C2 domain form intermolecular disulfide bonds. Using a modified tripartite split GFP complementation strategy and confocal microscopy, we also found that RAGE dimerization occurs in the endoplasmic reticulum (ER), and that RAGE mutant molecules without the double disulfide bridges are unstable, and are subjected to the ER-associated degradation. CONCLUSION Disulfide bond-mediated RAGE dimerization in the ER is the critical step of RAGE biogenesis. Without formation of intermolecular disulfide bonds in the C2 region, RAGE fails to reach cell surface. SIGNIFICANCE This is the first report of RAGE intermolecular disulfide bond.
Collapse
Affiliation(s)
- Wen Wei
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Leonie Lampe
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Sungha Park
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Bhavana S. Vangara
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Geoffrey S. Waldo
- Bioscience Division, MS-M888, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Stephanie Cabantous
- Bioscience Division, MS-M888, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Sarah S. Subaran
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Dongmei Yang
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Li Lin
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
22
|
Oxidative processing of latent Fas in the endoplasmic reticulum controls the strength of apoptosis. Mol Cell Biol 2012; 32:3464-78. [PMID: 22751926 DOI: 10.1128/mcb.00125-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We recently demonstrated that S-glutathionylation of the death receptor Fas (Fas-SSG) amplifies apoptosis (V. Anathy et al., J. Cell Biol. 184:241-252, 2009). In the present study, we demonstrate that distinct pools of Fas exist in cells. Upon ligation of surface Fas, a separate pool of latent Fas in the endoplasmic reticulum (ER) underwent rapid oxidative processing characterized by the loss of free sulfhydryl content (Fas-SH) and resultant increases in S-glutathionylation of Cys294, leading to increases of surface Fas. Stimulation with FasL rapidly induced associations of Fas with ERp57 and glutathione S-transferase π (GSTP), a protein disulfide isomerase and catalyst of S-glutathionylation, respectively, in the ER. Knockdown or inhibition of ERp57 and GSTP1 substantially decreased FasL-induced oxidative processing and S-glutathionylation of Fas, resulting in decreased death-inducing signaling complex formation and caspase activity and enhanced survival. Bleomycin-induced pulmonary fibrosis was accompanied by increased interactions between Fas-ERp57-GSTP1 and S-glutathionylation of Fas. Importantly, fibrosis was largely prevented following short interfering RNA-mediated ablation of ERp57 and GSTP. Collectively, these findings illuminate a regulatory switch, a ligand-initiated oxidative processing of latent Fas, that controls the strength of apoptosis.
Collapse
|
23
|
Anathy V, Roberson EC, Guala AS, Godburn KE, Budd RC, Janssen-Heininger YMW. Redox-based regulation of apoptosis: S-glutathionylation as a regulatory mechanism to control cell death. Antioxid Redox Signal 2012; 16:496-505. [PMID: 21929356 PMCID: PMC3304251 DOI: 10.1089/ars.2011.4281] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Redox-based signaling governs a number of important pathways in tissue homeostasis. Consequently, deregulation of redox-controlled processes has been linked to a number of human diseases. Among the biological processes regulated by redox signaling, apoptosis or programmed cell death is a highly conserved process important for tissue homeostasis. Apoptosis can be triggered by a wide variety of stimuli, including death receptor ligands, environmental agents, and cytotoxic drugs. Apoptosis has also been implicated in the etiology of many human diseases. RECENT ADVANCES Recent discoveries demonstrate that redox-based changes are required for efficient activation of apoptosis. Among these redox changes, alterations in the abundant thiol, glutathione (GSH), and the oxidative post-translational modification, protein S-glutathionylation (PSSG) have come to the forefront as critical regulators of apoptosis. CRITICAL ISSUES Although redox-based changes have been documented in apoptosis and disease pathogenesis, the mechanistic details, whereby redox perturbations intersect with pathogenic processes, remain obscure. FUTURE DIRECTIONS Further research will be needed to understand the context in which of the members of the death receptor pathways undergo ligand dependent oxidative modifications. Additional investigation into the interplay between oxidative modifications, redox enzymes, and apoptosis pathway members are also critically needed to improve our understanding how redox-based control is achieved. Such analyses will be important in understanding the diverse chronic diseases. In this review we will discuss the emerging paradigms in our current understanding of redox-based regulation of apoptosis with an emphasis on S-glutathionylation of proteins and the enzymes involved in this important post-translational modification.
Collapse
Affiliation(s)
- Vikas Anathy
- Department of Pathology, University of Vermont College of Medicine, Burlington, 05405, USA
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
During wound healing, contractile fibroblasts called myofibroblasts regulate the formation and contraction of granulation tissue; however, pathological and persistent myofibroblast activation, which occurs in hypertrophic scars or tissue fibrosis, results in a loss of function. Many reviews outline the cellular and molecular features of myofibroblasts and their roles in a variety of diseases. This review focuses on the origins of myofibroblasts and the factors that control their differentiation and prolonged survival in fibrotic tissues. Pulmonary fibrosis is used to illustrate many key points, but examples from other tissues and models are also included. Myofibroblasts originate mostly from tissue-resident fibroblasts, and also from epithelial and endothelial cells or other mesenchymal precursors. Their differentiation is influenced by cytokines, growth factors, extracellular matrix composition and stiffness, and cell surface molecules such as proteoglycans and THY1, among other factors. Many of these effects are modulated by cell contraction. Myofibroblasts resist programmed cell death, which promotes their accumulation in fibrotic tissues. The cause of resistance to apoptosis in myofibroblasts is under ongoing investigation, but many of the same stimuli that regulate their differentiation are involved. The contributions of oxidative stress, the WNT-β-catenin pathway and PPARγ to myofibroblast differentiation and survival are increasingly appreciated.
Collapse
|
25
|
Glutathione in cancer cell death. Cancers (Basel) 2011; 3:1285-310. [PMID: 24212662 PMCID: PMC3756414 DOI: 10.3390/cancers3011285] [Citation(s) in RCA: 225] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 02/22/2011] [Accepted: 03/09/2011] [Indexed: 01/08/2023] Open
Abstract
Glutathione (L-γ-glutamyl-L-cysteinyl-glycine; GSH) in cancer cells is particularly relevant in the regulation of carcinogenic mechanisms; sensitivity against cytotoxic drugs, ionizing radiations, and some cytokines; DNA synthesis; and cell proliferation and death. The intracellular thiol redox state (controlled by GSH) is one of the endogenous effectors involved in regulating the mitochondrial permeability transition pore complex and, in consequence, thiol oxidation can be a causal factor in the mitochondrion-based mechanism that leads to cell death. Nevertheless GSH depletion is a common feature not only of apoptosis but also of other types of cell death. Indeed rates of GSH synthesis and fluxes regulate its levels in cellular compartments, and potentially influence switches among different mechanisms of death. How changes in gene expression, post-translational modifications of proteins, and signaling cascades are implicated will be discussed. Furthermore, this review will finally analyze whether GSH depletion may facilitate cancer cell death under in vivo conditions, and how this can be applied to cancer therapy.
Collapse
|
26
|
Lyle PA, Mitsopoulos P, Suntres ZE. N-Acetylcysteine Modulates the Cytotoxic Effects of Paclitaxel. Chemotherapy 2011; 57:298-304. [DOI: 10.1159/000329510] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 02/18/2011] [Indexed: 11/19/2022]
|
27
|
Galan C, Jardín I, Dionisio N, Salido G, Rosado JA. Role of oxidant scavengers in the prevention of Ca²+ homeostasis disorders. Molecules 2010; 15:7167-87. [PMID: 20953160 PMCID: PMC6259185 DOI: 10.3390/molecules15107167] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/09/2010] [Accepted: 10/14/2010] [Indexed: 02/07/2023] Open
Abstract
A number of disorders, such as Alzheimer disease and diabetes mellitus, have in common the alteration of the redox balance, resulting in an increase in reactive oxygen species (ROS) generation that might lead to the development of apoptosis and cell death. It has long been known that ROS can significantly alter Ca²+ mobilization, an intracellular signal that is involved in the regulation of a wide variety of cellular functions. Cells have a limited capability to counteract the effects of oxidative stress, but evidence has been provided supporting the beneficial effects of exogenous ROS scavengers. Here, we review the effects of oxidative stress on intracellular Ca²+ homeostasis and the role of antioxidants in the prevention and treatment of disorders associated to abnormal Ca²+ mobilization induced by ROS.
Collapse
Affiliation(s)
| | | | | | | | - Juan A. Rosado
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34 927257139; Fax: +34 927257110
| |
Collapse
|