Role of vimentin in modulating immune cell apoptosis and inflammatory responses in sepsis

Longxiang Su, Pan Pan, Peng Yan, Yun Long, Xiang Zhou, Xiaoting Wang, Ruo Zhou, Bo Wen, Lixin Xie, Dawei Liu, Longxiang Su, Pan Pan, Peng Yan, Yun Long, Xiang Zhou, Xiaoting Wang, Ruo Zhou, Bo Wen, Lixin Xie, Dawei Liu

Abstract

New diagnostic biomarkers or therapeutic targets for sepsis have substantial significance for critical care medicine. In this study, 192 differentially expressed proteins were selected through iTRAQ. Based on cluster analysis of protein expression dynamics and protein-protein interactions, hemopexin, vimentin, and heat shock protein 90 were selected for further investigation. It was demonstrated that serum vimentin (VIM) levels were significantly increased in patients with sepsis and septic shock compared to controls and that VIM expression was significantly increased in lymphocytes isolated from septic shock and sepsis patients compared to controls. Moreover, a nonsurvivor group had higher serum VIM levels and VIM expression in lymphocytes. Caspase-3 was significantly upregulated in Jurkat T cells lacking VIM and when exposed to LPS compared to control cells. In contrast, caspase-3 was reduced nearly 40% in cells over-expressing VIM. IL-2, IL-10 and IFN-α levels were significantly decreased in cells lacking VIM compared to control cells, whereas they were not significantly altered in cells over-expressing VIM. These findings suggest that VIM modulates lymphocyte apoptosis and inflammatory responses and that VIM could be a new target for the diagnosis and prognostic prediction of patients with sepsis or septic shock.

Trial registration: ClinicalTrials.gov NCT01493466 NCT03253146.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Flowchart illustrating the clinical screening stage and clinical validation stage of the target proteins.
Figure 2
Figure 2
Protein expression patterns analyzed by series test of clusters. The expression patterns of 167 proteins were analyzed, and eight model profiles were used to summarize. Seven protein expression patterns showed significant p-values (p < 0.05). Inserts represent an identical expression trend for several column molecules. All of the molecules exhibiting this expression trend were displayed in different colored lines above the X-axis. Detailed information is shown in Supplemental Table 2.
Figure 3
Figure 3
Coexpression network of the proteins. Fifty-two proteins were analyzed and identified based on a protein coexpression network with k-core algorithm. Each circle represented one protein of potential interest. The size of each circle indicated the power of the interrelation among the proteins, and edges between two circles indicated interactions between proteins. The more edges of a given target, the more proteins connect to it and the more central role it plays within the network.
Figure 4
Figure 4
Comparison of serum vimentin concentrations and expression in lymphocytes. Serum vimentin (VIM) concentrations in sepsis patients and vimentin (VIM) expression in lymphocytes isolated from sepsis patients were quantified by ELISA and flow cytometry, respectively, as described in the Materials and Methods. Panel A,B: Comparison of serum vimentin (VIM) concentrations in the different groups by ELISA. Panel C,D: Representative dot plots and histograms of the flow cytometry analysis. Panel E,F: Comparison of vimentin (VIM) expression in lymphocytes isolated from control, sepsis and septic shock patients. *p < 0.05.
Figure 5
Figure 5
Vimentin expression in Jurkat cells and its effects on apoptosis. Jurkat cells were transfected with control siRNA, vimentin-specific siRNA or a plasmid expressing vimentin as described in the Materials and Methods. Cells were cultured in serum-free DMEM as indicated, followed by TUNEL assay analysis as described in the Materials and Methods. Panel A: Representative immunoblot indicating suppression of vimentin by siRNA (Vim-siRNA) or overexpression of vimentin (Vim-DNA). Panel B: Representative images of TUNEL assays in the absence of LPS. Green: TUNEL-positive staining; Red: cell nuclei.
Figure 6
Figure 6
Role of vimentin in modulating Jurkat cell apoptosis in response to LPS. Jurkat cells were transfected with control siRNA, vimentin-specific siRNA or a plasmid expressing vimentin as described in the Materials and Methods. Cells were then cultured for 6, 18 and 24 h in the presence or absence of LPS (10 µg/mL). Panel A: Representative images of TUNEL staining in the presence of LPS. Panel B: Quantitative comparison of TUNEL positivity in cells with or without LPS exposure. *p < 0.05 compared to control cells without LPS; #p < 0.05 compared to control cells with LPS. Data shown are from a single representative assessment; the experiments were repeated at least 3 times with similar results.
Figure 7
Figure 7
Expression of caspase-3 and Bcl-2 in Jurkat cells lacking vimentin or overexpressing vimentin. Jurkat cells were transfected with control siRNA, vimentin-specific siRNA or a plasmid expressing vimentin as described in the Materials and Methods. Cells were then cultured in serum-free DMEM in the presence or absence of LPS (10 µg/mL) for 24 h. Total cell lysates were analyzed via immunoblotting for caspase-3, Bcl-2 and ß-actin (as a loading control) as described in the Materials and Methods. Panel A: Representative immunoblots. Panel B: Semiquantitative comparison of target proteins with normalization. Vertical axes: protein level expressed as “Ratio versus control”, which was obtained as follows: (1). The densities of target protein versus internal control ß-actin were obtained. (2). The group of cells transfected with Con-siRNA and cultured in SF-DMEM was set as a reference (“Control = 1”), and the ratio of other groups versus this control group were obtained. Horizontal axes: cells transfected with control siRNA (Con-siRNA), vimentin-specific siRNA (Vim-siRNA) or vimentin expressing plasmid (Vim-vector). Open bar: cells were cultured in serum-free DMEM (SF-DMEM); closed bar: cells treated with 10 µg/mL LPS. #p < 0.05; *p < 0.05 compared to cells transfected with Con-siRNA and cultured in SF-DMEM. Data represent an average of 3 separate experiments.
Figure 8
Figure 8
Release of cytokines in the supernatants. Jurkat cells were transfected with control siRNA, vimentin-specific siRNA or a plasmid expressing vimentin as described in the Materials and Methods. Cells were then cultured in serum-free DMEM in the presence or absence of LPS (10 µg/mL) for 24 h. Supernatants were harvested and used for quantification of cytokines by ELISA as described in the Materials and Methods. Panel A: TNF-α. Panel B: IL-2. Panel C: IL-10. Panel D: IFN-γ. Vertical axes: cytokine level expressed as pg/day/105 cells; horizontal axes: cells transfected with control siRNA (Con-siRNA), vimentin-specific siRNA (Vim-siRNA) or the plasmid expressing vimentin (Vim-vector). Data are shown from one representative assessment; experiments were each repeated at least 3 times with similar results. Open bar: cells cultured in the serum-free DMEM (SF-DMEM); closed bar: cells treated with 10 µg/mL LPS. *p < 0.05 compared to cells cultured in SF-DMEM; #p < 0.05 compared to cells transfected with Con-siRNA and cultured in SF-DMEM.

References

    1. Singer M, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) JAMA. 2016;315:801–810. doi: 10.1001/jama.2016.0287.
    1. Martin GS. Sepsis, severe sepsis and septic shock: changes in incidence, pathogens and outcomes. Expert Rev Anti Infect Ther. 2012;10:701–706. doi: 10.1586/eri.12.50.
    1. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348:1546–1554. doi: 10.1056/NEJMoa022139.
    1. Moore JX, et al. Defining Sepsis Mortality Clusters in the United States. Crit Care Med. 2016;44:1380–1387. doi: 10.1097/CCM.0000000000001665.
    1. Zhou J, et al. Epidemiology and outcome of severe sepsis and septic shock in intensive care units in mainland China. PLoS One. 2014;9:e107181. doi: 10.1371/journal.pone.0107181.
    1. An G, Namas RA, Vodovotz Y. Sepsis: from pattern to mechanism and back. Crit Rev Biomed Eng. 2012;40:341–351. doi: 10.1615/CritRevBiomedEng.v40.i4.80.
    1. Dupuy AM, et al. Role of biomarkers in the management of antibiotic therapy: an expert panel review: I - currently available biomarkers for clinical use in acute infections. Ann Intensive Care. 2013;3:22. doi: 10.1186/2110-5820-3-22.
    1. Pierrakos C, Vincent JL. Sepsis biomarkers: a review. Crit Care. 2010;14:R15. doi: 10.1186/cc8872.
    1. Sandquist M, Wong HR. Biomarkers of sepsis and their potential value in diagnosis, prognosis and treatment. Expert Rev Clin Immunol. 2014;10:1349–1356. doi: 10.1586/1744666X.2014.949675.
    1. Su L, et al. Identification of novel biomarkers for sepsis prognosis via urinary proteomic analysis using iTRAQ labeling and 2D-LC-MS/MS. PLoS One. 2013;8:e54237. doi: 10.1371/journal.pone.0054237.
    1. Su LX, et al. Significance of low serum vitamin D for infection risk, disease severity and mortality in critically ill patients. Chin Med J (Engl) 2013;126:2725–2730.
    1. Su L, et al. Discrimination of sepsis stage metabolic profiles with an LC/MS-MS-based metabolomics approach. BMJ Open Respir Res. 2014;1:e000056. doi: 10.1136/bmjresp-2014-000056.
    1. Su L, et al. Dynamic changes in amino acid concentration profiles in patients with sepsis. PLoS One. 2015;10:e0121933. doi: 10.1371/journal.pone.0121933.
    1. Hsu PC, et al. Vimentin is involved in peptidylarginine deiminase 2-induced apoptosis of activated Jurkat cells. Mol Cells. 2014;37:426–434. doi: 10.14348/molcells.2014.2359.
    1. Moisan E, Girard D. Cell surface expression of intermediate filament proteins vimentin and lamin B1 in human neutrophil spontaneous apoptosis. J Leukoc Biol. 2006;79:489–498. doi: 10.1189/jlb.0405190.
    1. Lavastre V, Pelletier M, Saller R, Hostanska K, Girard D. Mechanisms involved in spontaneous and Viscum album agglutinin-I-induced human neutrophil apoptosis: Viscum album agglutinin-I accelerates the loss of antiapoptotic Mcl-1 expression and the degradation of cytoskeletal paxillin and vimentin proteins via caspases. J Immunol. 2002;168:1419–1427. doi: 10.4049/jimmunol.168.3.1419.
    1. Morishima N. Changes in nuclear morphology during apoptosis correlate with vimentin cleavage by different caspases located either upstream or downstream of Bcl-2 action. Genes Cells. 1999;4:401–414. doi: 10.1046/j.1365-2443.1999.00270.x.
    1. Byun Y, et al. Caspase cleavage of vimentin disrupts intermediate filaments and promotes apoptosis. Cell Death Differ. 2001;8:443–450. doi: 10.1038/sj.cdd.4400840.
    1. Bone RC, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101:1644–1655. doi: 10.1378/chest.101.6.1644.
    1. Levy MM, et al. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med. 2003;31:1250–1256. doi: 10.1097/01.CCM.0000050454.01978.3B.
    1. Dellinger RP, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Intensive Care Med. 2008;34:17–60. doi: 10.1007/s00134-007-0934-2.
    1. Diz AP, Truebano M, Skibinski DO. The consequences of sample pooling in proteomics: an empirical study. Electrophoresis. 2009;30:2967–2975. doi: 10.1002/elps.200900210.
    1. Ramoni MF, Sebastiani P, Kohane IS. Cluster analysis of gene expression dynamics. Proc Natl Acad Sci USA. 2002;99:9121–9126. doi: 10.1073/pnas.132656399.
    1. Miller LD, et al. Optimal gene expression analysis by microarrays. Cancer Cell. 2002;2:353–361. doi: 10.1016/S1535-6108(02)00181-2.
    1. Jansen R, Greenbaum D, Gerstein M. Relating whole-genome expression data with protein-protein interactions. Genome Res. 2002;12:37–46. doi: 10.1101/gr.205602.
    1. Li C, Li H. Network-constrained regularization and variable selection for analysis of genomic data. Bioinformatics. 2008;24:1175–1182. doi: 10.1093/bioinformatics/btn081.
    1. Wei Z, Li H. A Markov random field model for network-based analysis of genomic data. Bioinformatics. 2007;23:1537–1544. doi: 10.1093/bioinformatics/btm129.
    1. Zhang JD, Wiemann S. KEGGgraph: a graph approach to KEGG PATHWAY in R and bioconductor. Bioinformatics. 2009;25:1470–1471. doi: 10.1093/bioinformatics/btp167.
    1. Spirin V, Mirny LA. Protein complexes and functional modules in molecular networks. Proc Natl Acad Sci USA. 2003;100:12123–12128. doi: 10.1073/pnas.2032324100.
    1. Eriksson JE, et al. Introducing intermediate filaments: from discovery to disease. J Clin Invest. 2009;119:1763–1771. doi: 10.1172/JCI38339.
    1. Katsumoto T, Mitsushima A, Kurimura T. The role of the vimentin intermediate filaments in rat 3Y1 cells elucidated by immunoelectron microscopy and computer-graphic reconstruction. Biol Cell. 1990;68:139–146. doi: 10.1016/0248-4900(90)90299-I.
    1. Leader M, Collins M, Patel J, Henry K. Vimentin: an evaluation of its role as a tumour marker. Histopathology. 1987;11:63–72. doi: 10.1111/j.1365-2559.1987.tb02609.x.
    1. Mor-Vaknin N, et al. Murine colitis is mediated by vimentin. Sci Rep. 2013;3:1045. doi: 10.1038/srep01045.
    1. Mortensen JH, et al. Fragments of Citrullinated and MMP-degraded Vimentin and MMP-degraded Type III Collagen Are Novel Serological Biomarkers to Differentiate Crohn’s Disease from Ulcerative Colitis. J Crohns Colitis. 2015;9:863–872. doi: 10.1093/ecco-jcc/jjv123.
    1. Blaschek MA, et al. Relation of antivimentin antibodies to anticardiolipin antibodies in systemic lupus erythematosus. Ann Rheum Dis. 1988;47:708–716. doi: 10.1136/ard.47.9.708.
    1. Vasko R, et al. Vimentin fragments are potential markers of rheumatoid synovial fibroblasts. Clin Exp Rheumatol. 2016;34:513–520.
    1. Kinloch AJ, et al. In Situ Humoral Immunity to Vimentin in HLA-DRB1*03(+) Patients With Pulmonary Sarcoidosis. Front Immunol. 2018;9:1516. doi: 10.3389/fimmu.2018.01516.
    1. Li FJ, et al. Autoimmunity to Vimentin Is Associated with Outcomes of Patients with Idiopathic Pulmonary Fibrosis. J Immunol. 2017;199:1596–1605. doi: 10.4049/jimmunol.1700473.
    1. dos Santos G, et al. Vimentin regulates activation of the NLRP3 inflammasome. Nat Commun. 2015;6:6574. doi: 10.1038/ncomms7574.
    1. Thiagarajan PS, et al. Vimentin is an endogenous ligand for the pattern recognition receptor Dectin-1. Cardiovasc Res. 2013;99:494–504. doi: 10.1093/cvr/cvt117.
    1. Li Y, et al. Sepsis-induced elevation in plasma serotonin facilitates endothelial hyperpermeability. Sci Rep. 2016;6:22747. doi: 10.1038/srep22747.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13:260–268. doi: 10.1016/S1473-3099(13)70001-X.
    1. Condotta SA, Cabrera-Perez J, Badovinac VP, Griffith TS. T-cell-mediated immunity and the role of TRAIL in sepsis-induced immunosuppression. Crit Rev Immunol. 2013;33:23–40. doi: 10.1615/CritRevImmunol.2013006721.
    1. Castelino DJ, McNair P, Kay TW. Lymphocytopenia in a hospital population–what does it signify? Aust N Z J Med. 1997;27:170–174. doi: 10.1111/j.1445-5994.1997.tb00934.x.
    1. Cabrera-Perez J, Condotta SA, Badovinac VP, Griffith TS. Impact of sepsis on CD4 T cell immunity. J Leukoc Biol. 2014;96:767–777. doi: 10.1189/jlb.5MR0114-067R.
    1. Boomer JS, Green JM, Hotchkiss RS. The changing immune system in sepsis: is individualized immuno-modulatory therapy the answer? Virulence. 2014;5:45–56. doi: 10.4161/viru.26516.
    1. Lee SJ, Yoo JD, Choi SY, Kwon OS. The expression and secretion of vimentin in the progression of non-alcoholic steatohepatitis. BMB Rep. 2014;47:457–462. doi: 10.5483/BMBRep.2014.47.8.256.

Source: PubMed

3
Subscribe