1
|
Kim JE, Wang SH, Lee DS, Kim TH. Protein disulfide isomerase integrates toll-like receptor 4 and P2X7 receptor signaling pathways during lipopolysaccharide-induced neuroinflammation. Sci Rep 2025; 15:7906. [PMID: 40050375 PMCID: PMC11885452 DOI: 10.1038/s41598-025-92780-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 03/09/2025] Open
Abstract
P2X7 receptor (P2X7R) augments lipopolysaccharide (LPS)-toll-like receptor 4 (TLR4)-mediated neuroinflammation. These roles of P2X7R in neuroinflammation are relevant to nitrosative stress through nuclear factor-κB (NF-κB)-inducible nitric oxide synthase (iNOS) pathway, while the underlying mechanisms are largely unknown. In the present study, we investigated whether protein disulfide isomerase (PDI) is involved in the integration of TLR4-P2X7R functions in response to LPS in vivo. The present study showed that LPS elicited NF-κB-mediated PDI upregulation, iNOS induction and S-nitrosylated PDI (SNO-PDI) level, independent of S-nitrosylation of NF-κB p65 subunit, in P2X7R+/+ mice more than P2X7R-/- mice. SN50 (an NF-κB inhibitor) effectively diminished LPS-induced PDI upregulation in both P2X7R+/+ and P2X7R-/- mice. PDI knockdown attenuated LPS-induced p65 S276 phosphorylation and iNOS induction in both strains. Of interest, S-nitroso-N-acetyl-DL-penicillamine (SNAP, a NO donor) increased SNO-PDI level, surface P2X7R expression and p65 S276 phosphorylation in P2X7R+/+ mice under physiological condition. In P2X7R-/- mice, SNAP was less effective on NF-κB S276 phosphorylation, although SNO-PDI level was similar to that in P2X7R+/+ mice. Taken together, the present data demonstrate that PDI may be an intermediator to integrate TLR4- and P2X7R-mediated signaling pathways in a positive feedback loop, which would exert NF-κB-iNOS-mediated nitrosative stress during LPS-induced neuroinflammation.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea.
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, Korea.
| | - Su Hyeon Wang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, Korea
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, Korea
| |
Collapse
|
2
|
Souza-Smith FM, Molina PE, Maiya R. Chronic alcohol feeding alters lymph and plasma proteome in a rodent model. Life Sci 2023; 327:121818. [PMID: 37268288 PMCID: PMC10330892 DOI: 10.1016/j.lfs.2023.121818] [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: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Chronic alcohol consumption in rodents induces mesenteric collecting lymphatic vessel hyperpermeability, lymph leakage, and consequent immunometabolic dysregulation of the perilymphatic adipose tissue (PLAT). The specific lymphatic components mediating PLAT immunometabolic dysregulation remain to be identified. Specifically, whether alcohol impacts lymph composition is unknown. This study aimed to determine alcohol associated changes in lymph and plasma proteome. Adult male rats were fed a Lieber-DeCarli liquid diet containing 36 % of calories from alcohol for 10 weeks. Time-matched control animals were pair-fed. At sacrifice lymph was collected for 2 h using the lymph-fistula technique and plasma was collected prior to sacrifice. Quantitative discovery-based proteomics identified a total of 703 proteins. An integrative approach combining Ingenuity Pathway Analysis (IPA) and an unbiased network analysis using WGCNA (Weighted Gene Co-expression Network Analysis) was used to analyze the proteomics data. IPA results identified significant upregulation of a cluster of apolipoproteins in lymph from alcohol-fed animals compared with pair-fed controls and a downregulation of 34 proteins in the plasma from alcohol-fed animals. WGCNA analysis identified several candidate hub proteins in the lymph that were also significantly differentially expressed in lymph from alcohol-fed animals compared to that of pair-fed controls. WGCNA analysis of plasma identified a module without significant enrichment of differentially expressed proteins. Of the 59 proteins contained within this module, only 2 were significantly differentially expressed in plasma from alcohol-fed rats compared to plasma of pair-fed controls. Future studies will investigate further the functionality of the hub proteins affected by alcohol feeding in both lymph and plasma.
Collapse
Affiliation(s)
- Flavia M Souza-Smith
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA.
| | - Patricia E Molina
- Department of Physiology & Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA 70112, USA.
| | - Rajani Maiya
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
3
|
Xu X, Chiu J, Chen S, Fang C. Pathophysiological roles of cell surface and extracellular protein disulfide isomerase and their molecular mechanisms. Br J Pharmacol 2021; 178:2911-2930. [PMID: 33837960 DOI: 10.1111/bph.15493] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/23/2021] [Accepted: 04/04/2021] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase (PDI) is the prototypic member of the thiol isomerase family that catalyses disulfide bond rearrangement. Initially identified in the endoplasmic reticulum as folding catalysts, PDI and other members in its family have also been widely reported to reside on the cell surface and in the extracellular matrix. Although how PDI is exported and retained on the cell surface remains a subject of debate, this unique pool of PDI is developing into an important mechanism underlying the redox regulation of protein sulfhydryls that are critical for the cellular activities under various disease conditions. This review aims to provide an overview of the pathophysiological roles of surface and extracellular PDI and their underlying molecular mechanisms. Understanding the involvement of extracellular PDI in these diseases will advance our knowledge in the molecular aetiology to facilitate the development of novel pharmacological strategies by specifically targeting PDI in extracellular compartments.
Collapse
Affiliation(s)
- Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Joyce Chiu
- The Centenary Institute, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| |
Collapse
|
4
|
Shi Y, Wang C, Wu D, Zhu Y, Wang ZE, Peng X. Mechanistic study of PDIA1-catalyzed TFF3 dimerization during sepsis. Life Sci 2020; 255:117841. [PMID: 32454156 DOI: 10.1016/j.lfs.2020.117841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/01/2023]
Abstract
AIMS Trefoil factor 3 (TFF3) is a gut mucosal protective molecule that is secreted by intestinal goblet cells. The dimeric structure of TFF3 enables it to function in intestinal mucosal repair and to maintain its own stability. Protein disulfide isomerase a1 (PDIA1) can directly catalyze the formation, isomerization and reduction of disulfide bonds in proteins and may play an important role in the formation of TFF3 dimer. In this study, we focused on the specific molecular mechanism of TFF3 dimerization by PDIA1 and the changes during sepsis. METHODS We examined the changes of PDIA1 and TFF3 in sepsis rats and cell models and used a variety of experimental techniques to investigate the specific molecular mechanism of PDIA1-catalyzed TFF3 dimerization. KEY FINDINGS We found that PDIA1 can directly catalyze the dimerization of TFF3. Our MD model proposed that two TFF3 monomers form hydrogen bonds with the region b' of PDIA1 through two stepwise reactions. Furthermore, we propose that the Cys24-Cys27 active site at the region a' of PDIA1 mediates disulfide bond formation between the Cys79 residues of each of the two TFF3 monomers via deprotonation and nucleophilic attack. During sepsis, PDIA1 is downregulated and the excessive release of nitric oxide (NO) promoted PDIA1 nitrosylation. This modification reduced PDIA1 activity, which resulted in the corresponding decrease of TFF3 dimerization and compromised TFF3 dimer function. SIGNIFICANCE Our study revealed a novel mechanism for the inhibition of intestinal mucosal repair during sepsis and revealed novel targets for the prevention and treatment of sepsis.
Collapse
Affiliation(s)
- Yan Shi
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China; Institute of Trauma Orthopedic Surgery, The 920 Hospital of Joint Logistic Support Force of Chinese PLA, Kunming, Yunnan 650032, China
| | - Chao Wang
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Dan Wu
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Yuanjiao Zhu
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Zi-En Wang
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China; Department of Burns, Union Hospital, Fujian Medical University, Fuzhou 350001, China
| | - Xi Peng
- Clinical Medical Research Center, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Army Medical University, Chongqing 400038, China; Department of Burns, Union Hospital, Fujian Medical University, Fuzhou 350001, China; Shriners Burns Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America.
| |
Collapse
|
5
|
Rawarak N, Suttitheptumrong A, Reamtong O, Boonnak K, Pattanakitsakul SN. Protein Disulfide Isomerase Inhibitor Suppresses Viral Replication and Production during Antibody-Dependent Enhancement of Dengue Virus Infection in Human Monocytic Cells. Viruses 2019; 11:v11020155. [PMID: 30781856 PMCID: PMC6410196 DOI: 10.3390/v11020155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
One of several mechanisms that leads to the development of dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) is called antibody-dependent enhancement (ADE). Monocytes can be infected by the ADE phenomenon, which occurs in dengue secondary infection. This study aimed to investigate the proteins involved in ADE of DENV infection in the human monocytic cell line U937. The phosphoproteins were used to perform and analyze for protein expression using mass spectrometry (GeLC-MS/MS). The differential phosphoproteins revealed 1131 altered proteins compared between isotype- and DENV-specific antibody-treated monocytes. The altered proteins revealed 558 upregulated proteins and 573 downregulated proteins. Protein disulfide isomerase (PDI), which is an enzyme that had a high-ranking fold change and that catalyzes the formation, breakage, and rearrangement of disulfide bonds within a protein molecule, was selected for further study. PDI was found to be important for dengue virus infectivity during the ADE model. The effect of PDI inhibition was also shown to be involved in the early stage of life cycle by time-of-drug-addition assay. These results suggest that PDI is important for protein translation and virion assembly of dengue virus during infection in human monocytes, and it may play a significant role as a chaperone to stabilize dengue protein synthesis.
Collapse
Affiliation(s)
- Nantapon Rawarak
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Aroonroong Suttitheptumrong
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Kobporn Boonnak
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Sa-Nga Pattanakitsakul
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
6
|
Macur K, Grzenkowicz-Wydra J, Konieczna L, Bigda J, Temporini C, Tengattini S, Bączek T. A Proteomic-Based Approach to Study the Mechanism of Cytotoxicity Induced by Interleukin-1α and Cycloheximide. Chromatographia 2017; 81:47-56. [PMID: 29398714 PMCID: PMC5780535 DOI: 10.1007/s10337-017-3382-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/18/2017] [Accepted: 08/10/2017] [Indexed: 12/04/2022]
Abstract
Abstract The exposure of HeLa cells to interleukin-1 alpha (IL-1α) in the presence of cycloheximide (CHX) leads to the release of active tumor necrosis factor alpha (TNF-α), eliciting cytocidal effect on these cells. A mass spectrometry (MS)-based analysis of the qualitative proteomic profiles of the HeLa cells treated only with IL-1α, CHX or simultaneously with IL-1α and CHX, in comparison to an untreated control, enabled to distinguish protein candidates possibly involved in this process. Among them protein disulphide isomerase (PDI) seemed to be particularly interesting for further research. Therefore, we focused on quantitative changes of PDI levels in HeLa cells subjected to IL-1α and CHX. Enzyme-linked immunosorbent assay (ELISA) was employed for determination of PDI concentrations in the investigated, differently treated HeLa cells. The obtained results confirmed up-regulation of PDI only in the cells stimulated with IL-1α alone. In contrary, the PDI levels in HeLa cells exposed to both IL-1α and CHX, where apoptotic process was intensive, did not increase significantly. Finally, we discuss how different expression levels of PDI together with other proteins, which were detected in this study, may influence the induction of cytotoxic effect and modulate sensitivity to cytotoxic action of IL1. Graphical Abstract ![]()
Electronic supplementary material The online version of this article (doi:10.1007/s10337-017-3382-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katarzyna Macur
- 1Intercollegiate Faculty of Biotechnology UG and MUG, University of Gdańsk, ul. Abrahama 58, 80-307 Gdańsk, Poland
| | | | - Lucyna Konieczna
- 3Department of Pharmaceutical Chemistry, Medical University of Gdańsk, al. Hallera 107, 80-416 Gdańsk, Poland
| | - Jacek Bigda
- 4Cell Biology Unit, Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG and MUG, Medical University of Gdańsk, ul. Dębinki 1, 80-210 Gdańsk, Poland
| | - Caterina Temporini
- 5Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Sara Tengattini
- 5Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Tomasz Bączek
- 3Department of Pharmaceutical Chemistry, Medical University of Gdańsk, al. Hallera 107, 80-416 Gdańsk, Poland
| |
Collapse
|
7
|
Vandooren J, Swinnen W, Ugarte-Berzal E, Boon L, Dorst D, Martens E, Opdenakker G. Endotoxemia shifts neutrophils with TIMP-free gelatinase B/MMP-9 from bone marrow to the periphery and induces systematic upregulation of TIMP-1. Haematologica 2017; 102:1671-1682. [PMID: 28775117 PMCID: PMC5622851 DOI: 10.3324/haematol.2017.168799] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/27/2017] [Indexed: 01/02/2023] Open
Abstract
Lipopolysaccharides or endotoxins elicit an excessive host inflammatory response and lead to life-threatening conditions such as endotoxemia and septic shock. Lipopolysaccharides trigger mobilization and stimulation of leukocytes and exaggerated production of pro-inflammatory molecules including cytokines and proteolytic enzymes. Matrix metalloproteinase-9 (MMP-9) or gelatinase B, a protease stored in the tertiary granules of polymorphonuclear leukocytes, has been implicated in such inflammatory reactions. Moreover, several studies even pinpointed MMP-9 as a potential target molecule to counter excessive inflammation in endotoxemia. Whereas the early effect of lipopolysaccharide-induced inflammation in vivo on the expression of MMP-9 in various peripheral organs has been described, the effects on the bone marrow and during late stage endotoxemia remain elusive. We demonstrate that TIMP-free MMP-9 is a major factor in bone marrow physiology and pathology. By using a mouse model for late-stage endotoxemia, we show that lipopolysaccharides elicited a depletion of neutrophil MMP-9 in the bone marrow and a shift of MMP-9 and MMP-9-containing cells towards peripheral organs, a pattern which was primarily associated with a relocation of CD11bhighGr-1high cells. In contrast, analysis of the tissue inhibitors of metalloproteinases was in line with a natural, systematic upregulation of TIMP-1, the main tissue inhibitor of TIMP-free MMP-9, and a general shift toward control of matrix metalloproteinase activity by tissue inhibitors of metalloproteinases.
Collapse
Affiliation(s)
- Jennifer Vandooren
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Wannes Swinnen
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Lise Boon
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Daphne Dorst
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Erik Martens
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Belgium.
| |
Collapse
|
8
|
Peichoto ME, Santoro ML. Patagonfibrase modifies protein expression of tissue factor and protein disulfide isomerase in rat skin. Toxicon 2016; 119:330-5. [PMID: 27390042 DOI: 10.1016/j.toxicon.2016.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/01/2016] [Indexed: 01/27/2023]
Abstract
Patagonfibrase is a hemorrhagic metalloproteinase isolated from the venom of the South American rear-fanged snake Philodryas patagoniensis, and is an important contributor to local lesions inflicted by this species. The tissue factor (TF)-factor VIIa complex, besides triggering the coagulation cascade, has been demonstrated to be involved in inflammatory events. Our aim was to determine whether patagonfibrase affects the expression of TF and protein disulfide isomerase (PDI), an enzyme that controls TF biological activity, at the site of patagonfibrase injection, and thus if they may play a role in hemostatic and inflammatory events induced by snake venoms. Patagonfibrase (60 μg/kg) was administered s.c. to rats, and after 3 h blood was collected to evaluate hemostasis parameters, and skin fragments close to the site of injection were taken to assess TF and PDI expression. Patagonfibrase did not alter blood cell counts, plasma fibrinogen levels, or levels of TF activity in plasma. However, by semiquantitative Western blotting, patagonfibrase increased TF expression by 2-fold, and decreased PDI expression by 3-fold in skin samples. In agreement, by immunohistochemical analyses, prominent TF expression was observed in the subcutaneous tissue. Thus, patagonfibrase affects the local expression of TF and PDI without inducing any systemic hemostatic disturbance, although that they may be involved in the local inflammatory events induced by hemorrhagic metalloproteinases. Once antivenom therapy is not totally effective to treat the local injury induced by snake venoms, modulation of the activity and expression of TF and/or PDI might become a strategy for treating snake envenomation.
Collapse
Affiliation(s)
- María Elisa Peichoto
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Instituto Nacional de Medicina Tropical, Neuquén y Jujuy s/n, 3370 Puerto Iguazú, Argentina.
| | - Marcelo Larami Santoro
- Laboratório de Fisiopatologia, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900 São Paulo, SP, Brazil.
| |
Collapse
|
9
|
Zhou J, Yang F, Zhou L, Wang JG, Wen P, Luo H, Li W, Song Z, Sharman E, Bondy S. Dietary melatonin attenuates age-related changes in morphology and in levels of key proteins in globus pallidus of mouse brain. Brain Res 2014; 1546:1-8. [DOI: 10.1016/j.brainres.2013.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 10/08/2013] [Accepted: 12/11/2013] [Indexed: 01/26/2023]
|
10
|
Grek C, Townsend D. Protein Disulfide Isomerase Superfamily in Disease and the Regulation of Apoptosis. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2014; 1:4-17. [PMID: 25309899 PMCID: PMC4192724 DOI: 10.2478/ersc-2013-0001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular homeostasis requires the balance of a multitude of signaling cascades that are contingent upon the essential proteins being properly synthesized, folded and delivered to appropriate subcellular locations. In eukaryotic cells the endoplasmic reticulum (ER) is a specialized organelle that is the central site of synthesis and folding of secretory, membrane and a number of organelletargeted proteins. The integrity of protein folding is enabled by the presence of ATP, Ca++, molecular chaperones, as well as an oxidizing redox environment. The imbalance between the load and capacity of protein folding results in a cellular condition known as ER stress. Failure of these pathways to restore ER homeostasis results in the activation of apoptotic pathways. Protein disulfide isomerases (PDI) compose a superfamily of oxidoreductases that have diverse sequences and are localized in the ER, nucleus, cytosol, mitochondria and cell membrane. The PDI superfamily has multiple functions including, acting as molecular chaperones, protein-binding partners, and hormone reservoirs. Recently, PDI family members have been implicated in the regulation of apoptotic signaling events. The complexities underlying the molecular mechanisms that define the switch from pro-survival to pro-death response are evidenced by recent studies that reveal the roles of specific chaperone proteins as integration points in signaling pathways that determine cell fate. The following review discusses the dual role of PDI in cell death and survival during ER stress.
Collapse
Affiliation(s)
- C. Grek
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - D.M. Townsend
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
11
|
Camargo LDL, Babelova A, Mieth A, Weigert A, Mooz J, Rajalingam K, Heide H, Wittig I, Lopes LR, Brandes RP. Endo-PDI is required for TNFα-induced angiogenesis. Free Radic Biol Med 2013; 65:1398-1407. [PMID: 24103565 DOI: 10.1016/j.freeradbiomed.2013.09.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/08/2013] [Accepted: 09/29/2013] [Indexed: 12/20/2022]
Abstract
Protein disulfide isomerase (PDI) and its homologs are oxidoreductases facilitating protein folding in the ER. Endo-PDI (also termed ERp46) is highly expressed in endothelial cells. It belongs to the PDI family but its physiological function is largely unknown. We studied the role of Endo-PDI in endothelial angiogenic responses. Stimulation of human umbilical vein endothelial cells (with TNFα (10ng/ml) increased ERK1/2 phosphorylation. This effect was largely attenuated by Endo-PDI siRNA, whereas JNK and p38 MAP kinase phosphorylation was Endo-PDI independent. Similarly, TNFα-stimulated NF-κB signaling determined by IκBα degradation as well as TNFα-induced ICAM expression was unaffected by Endo-PDI siRNA. The action of Endo-PDI was not mediated by extracellular thiol exchange or cell surface PDI as demonstrated by nonpermeative inhibitors and PDI-neutralizing antibody. Moreover, exogenously added PDI failed to restore ERK1/2 activation after Endo-PDI knockdown. This suggests that Endo-PDI acts intracellularly potentially by maintaining the Ras/Raf/MEK/ERK pathway. Indeed, knockdown of Endo-PDI attenuated Ras activation measured by G-LISA and Raf phosphorylation. ERK activation influences gene expression by the transcriptional factor AP-1, which controls MMP-9 and cathepsin B, two proteases required for angiogenesis. TNFα-stimulated MMP-9 and cathepsin B induction was reduced by silencing of Endo-PDI. Accordingly, inhibition of cathepsin B or Endo-PDI siRNA blocked the TNFα-stimulated angiogenic response in the spheroid outgrowth assays. Moreover ex vivo tube formation and in vivo Matrigel angiogenesis in response to TNFα were attenuated by Endo-PDI siRNA. In conclusion, our study establishes Endo-PDI as a novel, important mediator of AP-1-driven gene expression and endothelial angiogenic function.
Collapse
Affiliation(s)
- Livia de Lucca Camargo
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany; Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrea Babelova
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Anja Mieth
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute for Biochemistry I, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Juliane Mooz
- Institute for Biochemistry II, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | | | - Heinrich Heide
- Functional Proteomics, SFB815 Core Unit, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB815 Core Unit, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Lucia Rossetti Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
12
|
Protective effects of polydatin on septic lung injury in mice via upregulation of HO-1. Mediators Inflamm 2013; 2013:354087. [PMID: 23431240 PMCID: PMC3570923 DOI: 10.1155/2013/354087] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/02/2013] [Accepted: 01/02/2013] [Indexed: 11/17/2022] Open
Abstract
The present study was carried out to investigate the effects and mechanisms of polydatin (PD) in septic mice. The model of cecal ligation and puncture (CLP-)induced sepsis was employed. Pretreatment of mice with PD (15, 45, and 100 mg/kg) dose-dependently reduced sepsis-induced mortality and lung injury, as indicated by alleviated lung pathological changes and infiltration of proteins and leukocytes. In addition, PD inhibited CLP-induced serum tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) production, lung cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase isoform (iNOS) protein expressions and NF-κB activation. Notably, PD upregulated the expression and activity of heme oxygenase (HO-)1 in lung tissue of septic mice. Further, the protective effects of PD on sepsis were abrogated by ZnPP IX, a specific HO-1 inhibitor. These findings indicated that PD might be an effective antisepsis drug.
Collapse
|
13
|
Pescatore LA, Bonatto D, Forti FL, Sadok A, Kovacic H, Laurindo FRM. Protein disulfide isomerase is required for platelet-derived growth factor-induced vascular smooth muscle cell migration, Nox1 NADPH oxidase expression, and RhoGTPase activation. J Biol Chem 2012; 287:29290-300. [PMID: 22773830 DOI: 10.1074/jbc.m112.394551] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Vascular Smooth Muscle Cell (VSMC) migration into vessel neointima is a therapeutic target for atherosclerosis and postinjury restenosis. Nox1 NADPH oxidase-derived oxidants synergize with growth factors to support VSMC migration. We previously described the interaction between NADPH oxidases and the endoplasmic reticulum redox chaperone protein disulfide isomerase (PDI) in many cell types. However, physiological implications, as well as mechanisms of such association, are yet unclear. We show here that platelet-derived growth factor (PDGF) promoted subcellular redistribution of PDI concomitant to Nox1-dependent reactive oxygen species production and that siRNA-mediated PDI silencing inhibited such reactive oxygen species production, while nearly totally suppressing the increase in Nox1 expression, with no change in Nox4. Furthermore, PDI silencing inhibited PDGF-induced VSMC migration assessed by distinct methods, whereas PDI overexpression increased spontaneous basal VSMC migration. To address possible mechanisms of PDI effects, we searched for PDI interactome by systems biology analysis of physical protein-protein interaction networks, which indicated convergence with small GTPases and their regulator RhoGDI. PDI silencing decreased PDGF-induced Rac1 and RhoA activities, without changing their expression. PDI co-immunoprecipitated with RhoGDI at base line, whereas such association was decreased after PDGF. Also, PDI co-immunoprecipitated with Rac1 and RhoA in a PDGF-independent way and displayed detectable spots of perinuclear co-localization with Rac1 and RhoGDI. Moreover, PDI silencing promoted strong cytoskeletal changes: disorganization of stress fibers, decreased number of focal adhesions, and reduced number of RhoGDI-containing vesicular recycling adhesion structures. Overall, these data suggest that PDI is required to support Nox1/redox and GTPase-dependent VSMC migration.
Collapse
Affiliation(s)
- Luciana A Pescatore
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil 05403-000
| | | | | | | | | | | |
Collapse
|
14
|
Pierrard MA, Kestemont P, Phuong NT, Tran MP, Delaive E, Thezenas ML, Dieu M, Raes M, Silvestre F. Proteomic analysis of blood cells in fish exposed to chemotherapeutics: evidence for long term effects. J Proteomics 2012; 75:2454-67. [PMID: 22406376 DOI: 10.1016/j.jprot.2012.02.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/10/2012] [Accepted: 02/17/2012] [Indexed: 01/03/2023]
Abstract
Proteomics technology are increasingly used in ecotoxicological studies to characterize and monitor biomarkers of exposure. The present study aims at identifying long term effects of malachite green (MG) exposure on the proteome of peripheral blood mononuclear cells (PBMC) from the Asian catfish, Pangasianodon hypophthalmus. A common (0.1 ppm) concentration for therapeutic treatment was applied twice with a 72 h interval. PBMC were collected directly at the end of the second bath of MG (T1) and after 1 month of decontamination (T2). Analytical 2D-DIGE gels were run and a total of 2551±364 spots were matched. Among them, MG induced significant changes in abundance of 116 spots with no recovery after one month of decontamination. Using LC-MS/MS and considering single identification per spot, we could identify 25 different proteins. Additionally, MG residues were measured in muscle and in blood indicating that leuco-MG has almost totally disappeared after one month of decontamination. This work highlights long term effects of MG treatment on the PBMC proteome from fish intended for human consumption.
Collapse
Affiliation(s)
- Marie-Aline Pierrard
- Research Unit in Environmental and Evolutionary Biology (URBE), University of Namur (FUNDP), Rue de Bruxelles 61, B-5000, Namur, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mishra K, Shweta, Diwaker D, Ganju L. Dengue virus infection induces upregulation of hn RNP-H and PDIA3 for its multiplication in the host cell. Virus Res 2012; 163:573-9. [DOI: 10.1016/j.virusres.2011.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 11/30/2022]
|
16
|
Mallawaaratchy DM, Mactier S, Kaufman KL, Blomfield K, Christopherson RI. The phosphoinositide 3-kinase inhibitor LY294002, decreases aminoacyl-tRNA synthetases, chaperones and glycolytic enzymes in human HT-29 colorectal cancer cells. J Proteomics 2011; 75:1590-9. [PMID: 22172953 DOI: 10.1016/j.jprot.2011.11.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 11/03/2011] [Accepted: 11/25/2011] [Indexed: 01/10/2023]
Abstract
The proposed anticancer drug LY294002, inhibits phosphoinositide-3 kinase (PI3K) that initiates a signalling pathway often activated in colorectal cancer (CRC). The effects of LY294002 (10 μM, 48 h) on the cytosolic, mitochondrial and nuclear proteomes of human HT-29 CRC cells have been determined using iTRAQ (isobaric tag for relative and absolute quantitation) and tandem mass spectrometry (MS/MS). Analysis of cells treated with LY294002 identified 26 differentially abundant proteins that indicate several mechanisms of action. The majority of protein changes were directly or indirectly associated with Myc and TNF-α, previously implicated in CRC progression. LY294002 decreased the levels of 6 aminoacyl-tRNA synthetases (average 0.39-fold) required for protein translation, 5 glycolytic enzymes (average 0.37-fold) required for ATP synthesis, and 3 chaperones required for protein folding. There was a 3.2-fold increase in lysozyme C involved in protein-glycoside hydrolysis. LY294002 increased cytosolic p53 with a concomitant decrease in nuclear p53, suggesting transfer of p53 to the cytosol where apoptosis might be initiated via the intrinsic mitochondrial pathway. Protein changes described here suggest that the anti-angiogenic effects of LY294002 may be related to p53; the mutational status of p53 in CRC may be an important determinant of the efficacy of PI3K inhibitors for treatment.
Collapse
|
17
|
Wei SD, Li JZ, Liu ZJ, Chen Q, Chen Y, Chen M, Gong JP. Dexamethasone attenuates lipopolysaccharide-induced liver injury by downregulating glucocorticoid-induced tumor necrosis factor receptor ligand in Kupffer cells. Hepatol Res 2011; 41:989-99. [PMID: 21951872 DOI: 10.1111/j.1872-034x.2011.00852.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AIM Glucocorticoid-induced tumor necrosis factor receptor ligand (GITRL) plays pro-inflammatory roles in immune response. Thus, our aim was to assess if dexamethasone attenuates lipopolysaccharide (LPS)-induced liver injury by affecting GITRL in Kupffer cells (KC). METHODS A BALB/c mouse model of liver injury was established by i.p. injecting with LPS (10 mg/kg) co-treated with or without dexamethasone (3 mg/kg). Blood and liver samples were obtained for analysis of liver morphology, GITRL expression, hepatocellular function and cytokine levels at 24 h after injection. KC were isolated and challenged by LPS (1 µg/mL), with or without dexamethasone (10 µM) co-treatment, or with GITRL siRNA pre-transfection. The GITRL expression and cytokine levels were assayed at 24 h after challenge. RESULTS Dexamethasone treatment significantly improved the survival rate of endotoxemic mice (P < 0.05), whereas serum alanine aminotransferase, aspartate aminotransferase, tumor necrosis factor (TNF)-α, interleukin (IL)-6 and γ-interferon levels were significantly decreased (P < 0.05, respectively). Concurrently, LPS-induced hepatic tissue injury was attenuated as indicated by morphological analysis; and expression of GITRL in liver tissue and KC was downregulated (P < 0.05). Consistent with these in vivo experiments, inhibited expression of GITRL, TNF-α and IL-6 caused by dexamethasone treatment were also observed in LPS-stimulated KC. The GITRL, TNF-α and IL-6 expression was also significantly inhibited by GITRL gene silencing. CONCLUSION The TNF-α and IL-6 expression of LPS-stimulated KC was inhibited by GITRL gene silencing. Dexamethasone attenuates LPS-induced liver injury, at least proportionately, by downregulating GITRL in KC.
Collapse
Affiliation(s)
- Si D Wei
- Key Laboratory of Chongqing Hepatobiliary Surgery Key Laboratory of Molecular Biology for Infectious Diseases, People's Republic of China Ministry of Education, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Severino V, Chambery A, Vitiello M, Cantisani M, Galdiero S, Galdiero M, Malorni L, Di Maro A, Parente A. Proteomic Analysis of Human U937 Cell Line Activation Mediated by Haemophilus influenzae Type b P2 Porin and Its Surface-Exposed Loop 7. J Proteome Res 2010; 9:1050-62. [DOI: 10.1021/pr900931n] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Valeria Severino
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Angela Chambery
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Mariateresa Vitiello
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Marco Cantisani
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Stefania Galdiero
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Massimiliano Galdiero
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Livia Malorni
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Antimo Di Maro
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| | - Augusto Parente
- Department of Life Science, Second University of Naples, Via Vivaldi 43, I-81100 Caserta, Italy, Department of Experimental Medicine, Second University of Naples, Via De Crecchio 7, I-80138 Napoli, Italy, Department of Biological Sciences, University of Naples “Federico II”, Via Mezzocannone 16, I-80134, Napoli, Italy, Institute of Biostructure and Bioimaging, C.N.R., Via Mezzocannone 16, I-80134, Napoli, Italy, and Proteomic and Biomolecular Mass Spectrometry Center, Institute of Food Science and
| |
Collapse
|
19
|
Opal SM, LaRosa SP. Year in review 2008: Critical Care--sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2009; 13:224. [PMID: 19886974 PMCID: PMC2784335 DOI: 10.1186/cc7945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The present report highlights the most important papers appearing in Critical Care and other major journals about severe sepsis, the systemic inflammatory response and multiorgan dysfunction over the past year. A number of these clinical and laboratory studies will have a considerable impact on the sepsis research agenda for years to come. The steroid controversy, the debate over tight glycemic control, the colloid versus crystalloid issue, the value of selective decontamination of the digestive tract, the enlarging role of biomarkers, the value of genomics and rapid diagnostic techniques have all been prominently featured in recent publications. Basic research into novel predictive assays, genetic polymorphisms, and new molecular methods to risk-stratify and to determine treatment options for sepsis have occupied much of the Critical Care publications relating to sepsis pathophysiology in 2008. We will attempt to briefly summarize what we consider to be the most significant contributions to the sepsis literature over the last year, and their likely ramifications in the future, for critical care clinicians, clinical investigators and basic researchers alike.
Collapse
Affiliation(s)
- Steven M Opal
- Department of Medicine, Infectious Disease Division, Memorial Hospital of RI, The Warren Alpert Medical School of Brown University, 111 Brewster Street, Pawtucket, RI 02860, USA.
| | | |
Collapse
|
20
|
Kim Y, Kang K, Kim I, Lee YJ, Oh C, Ryoo J, Jeong E, Ahn K. Molecular mechanisms of MHC class I-antigen processing: redox considerations. Antioxid Redox Signal 2009; 11:907-36. [PMID: 19178136 DOI: 10.1089/ars.2008.2316] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Major histocompatibility complex (MHC) class I molecules present antigenic peptides to the cell surface for screening by CD8(+) T cells. A number of ER-resident chaperones assist the assembly of peptides onto MHC class I molecules, a process that can be divided into several steps. Early folding of the MHC class I heavy chain is followed by its association with beta(2)-microglobulin (beta(2)m). The MHC class I heavy chain-beta(2)m heterodimer is incorporated into the peptide-loading complex, leading to peptide loading, release of the peptide-filled MHC class I molecules from the peptide-loading complex, and exit of the complete MHC class I complex from the ER. Because proper antigen presentation is vital for normal immune responses, the assembly of MHC class I molecules requires tight regulation. Emerging evidence indicates that thiol-based redox regulation plays critical roles in MHC class I-restricted antigen processing and presentation, establishing an unexpected link between redox biology and antigen processing. We review the influences of redox regulation on antigen processing and presentation. Because redox signaling pathways are a rich source of validated drug targets, newly discovered redox biology-mediated mechanisms of antigen processing may facilitate the development of more selective and therapeutic drugs or vaccines against immune diseases.
Collapse
Affiliation(s)
- Youngkyun Kim
- National Creative Research Center for Antigen Presentation, Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|