Proteomics and metabonomics analyses of Covid-19 complications in patients with pulmonary fibrosis

Jianrong Yang, Chunxia Chen, Wan Chen, Luying Huang, Zhao Fu, Kun Ye, Liwen Lv, Zhihuang Nong, Xing Zhou, Wensheng Lu, Mei Zhong, Jianrong Yang, Chunxia Chen, Wan Chen, Luying Huang, Zhao Fu, Kun Ye, Liwen Lv, Zhihuang Nong, Xing Zhou, Wensheng Lu, Mei Zhong

Abstract

Pulmonary fibrosis is a devastating disease, and the pathogenesis of this disease is not completely clear. Here, the medical records of 85 Covid-19 cases were collected, among which fibrosis and progression of fibrosis were analyzed in detail. Next, data independent acquisition (DIA) quantification proteomics and untargeted metabolomics were used to screen disease-related signaling pathways through clustering and enrichment analysis of the differential expression of proteins and metabolites. The main imaging features were lesions located in the bilateral lower lobes and involvement in five lobes. The closed association pathways were FcγR-mediated phagocytosis, PPAR signaling, TRP-inflammatory pathways, and the urea cycle. Our results provide evidence for the detection of serum biomarkers and targeted therapy in patients with Covid-19.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Figure 1
Figure 1
Representative chest CT images. (a) Ground-glass opacities in the dorsal segment of the left lower lobe of lung were seen in a 44-year-old female on admission, and partial absorption of lung lesions before discharge (b), Multiple fibrotic stripes shadows in dorsal segment of the lower lobe of the right lung were seen in a 62-year-old male after symptom onset (c), however, no obvious changes were observed before discharge (d, e), multiple fibrotic stripes shadows and patchy dense shadow (exudative lesion) were shown in a 65-year-old severe male on admission. (f) The exudative lesions were less and the fibrosis lesions were more than before in 20 days after hospitalization.
Figure 2
Figure 2
Differentially expressed proteins (DEPs) were identified and clustered in Covid-19 patients with pulmonary fibrosis. (a) Volcano plots of total protein levels. Red dots, significantly upregulated proteins; green dots, significantly downregulated proteins. (b) Cluster chart of DEPs. Red rows, significantly upregulated proteins; blue rows present significantly downregulated proteins.
Figure 3
Figure 3
Quality control of samples in Covid-19 patients with pulmonary fibrosis. (a, b) Coefficient of variation (CV) plots for positive and negative ion modes. (c,d) PLS-DA model plots for positive and negative ion modes. A, Covid-19 patients without pulmonary fibrosis (n = 6). B, Covid-19 patients with pulmonary fibrosis (n = 22).
Figure 4
Figure 4
Positive differentially expressed metabolites were identified, clustered and enriched in Covid-19 patients with pulmonary fibrosis. (a) Volcano plots of positive compounds. Red dots, significantly upregulated metabolites; green dots, significantly downregulated metabolites. (b) Heatmap of different positive metabolites (“pheatmap” package in R software v3.5.0, https://www.r-project.org/). Each row denotes a different metabolite, and each column denotes a sample. A, Covid-19 patients without pulmonary fibrosis (n = 6). B, Covid-19 patients with pulmonary fibrosis (n = 22). (c) Bubble plot of KEGG enrichment of positive metabolic pathways. The size of the circle dot denotes the number of different metabolites. RichFactor is defined as the number of differential metabolites annotated to the pathway divided by all identified metabolites annotated to the pathway.
Figure 5
Figure 5
Differentially expressed proteins (DEPs) were identified and analyzed using proteomics in Covid-19 patients with progressive pulmonary fibrosis. (a) Volcano plots of total protein level. (b) Cluster chart of DEPs. (c) Enrichment analysis of DEPs by GO function. (d) Bubble plot of the top 20 KEGG enrichment pathways of DEPs. RichFactor is defined as the number of differential metabolites annotated to the pathways divided by all identified metabolites annotated to the pathway. (e) Pathway network of DEPs (“igraph” package in R software v3.5.0, https://www.r-project.org/). The red and blue dots represent up-regulated and down-regulated differential proteins, respectively. The purple balls represent the top 10 pathways of enrichment. The area is directly proportional to the enrichment degree.
Figure 6
Figure 6
Positive differentially expressed metabolites were identified and analyzed using proteomics in Covid-19 patients with progressive pulmonary fibrosis. (a) PLS-DA model plot for positive ion mode. (b) Volcano plots of positive compounds. (c) Heatmap of different positive metabolites (“pheatmap” package in R software v3.5.0, https://www.r-project.org/). (d) Bubble plot of the top 20 KEGG enrichment of positive metabolic pathways. C, Nonprogressive pulmonary fibrosis of Covid-19 patients. D, Progressive pulmonary fibrosis of Covid-19 patients. RichFactor is defined as the number of differential metabolites annotated to the pathway divided by all identified metabolites annotated to the pathway.
Figure 7
Figure 7
Association analysis of proteomics and metabolomics in Covid-19 patients with progressive pulmonary fibrosis. (a) Cluster heat map of the correlation between proteomics and metabolomics (“pheatmap” package in R software v3.5.0, https://www.r-project.org/). Each row denotes a differentially expressed metabolite, and each column denote a DEP. Blue, negative correlation; Red, positive correlation. (b) Concentric circles of correlation between differential proteins and differential metabolites. If the angle between a differential protein and differential metabolite is an acute angle (less than 90 degrees), the correlation is positive. If the angle between the differential protein and the differential metabolite is an oblique angle (greater than 90 degrees and less than 180 degrees), the correlation is negative. Starting from the center of the circle, the lines are connected to the differential metabolites and differential proteins. The longer the connection length, the stronger the relationship.

References

    1. Wu Y, et al. SARS-CoV-2 is an appropriate name for the new coronavirus. Lancet. 2020;395:949–950. doi: 10.1016/S0140-6736(20)30557-2.
    1. WHO. Novel coronavirus disease named COVID-19. . Accessed 11 Feb 2020.
    1. WHO. Coronavirus disease (COVID-19) weekly epidemiological update and weekly operational update. . Accessed 25 Oct 2020.
    1. Lurie N, Saville M, Hatchett R, Halton J. Developing Covid-19 vaccines at pandemic speed. N. Engl. J. Med. 2020;382:1969–1973. doi: 10.1056/NEJMp2005630.
    1. Zhu FC, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet. 2020;395:1845–1854. doi: 10.1016/S0140-6736(20)31208-3.
    1. Xu YH, et al. Clinical and computed tomographic imaging features of novel coronavirus pneumonia caused by SARS-CoV-2. J. Infect. 2020;80:394–400. doi: 10.1016/j.jinf.2020.02.017.
    1. Bradley BT, et al. Histopathology and ultrastructural findings of fatal COVID-19 infections in Washington State: a case series. Lancet. 2020;396:320–332. doi: 10.1016/S0140-6736(20)31305-2.
    1. George PM, Patterson CM, Reed AK, Thillai M. Lung transplantation for idiopathic pulmonary fibrosis. Lancet Respir. Med. 2019;7:271–282. doi: 10.1016/S2213-2600(18)30502-2.
    1. Li F. Receptor recognition mechanisms of coronaviruses: a decade of structural studies. J. Virol. 2015;89:1954–1964. doi: 10.1128/JVI.02615-14.
    1. Qiao B, de la Olvera CM. Enhanced binding of SARS-CoV-2 spike protein to receptor by distal polybasic cleavage sites. ACS Nano. 2020;14:10616–10623. doi: 10.1021/acsnano.0c04798.
    1. Ganter MT, et al. Interleukin-1beta causes acute lung injury via alphavbeta5 and alphavbeta6 integrin-dependent mechanisms. Circ. Res. 2008;102:804–812. doi: 10.1161/CIRCRESAHA.107.161067.
    1. Shimbori C, et al. Mechanical stress-induced mast cell degranulation activates TGF-β1 signalling pathway in pulmonary fibrosis. Thorax. 2019;74:455–465. doi: 10.1136/thoraxjnl-2018-211516.
    1. Li M, et al. Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis. J. Clin. Invest. 2011;121:277–287. doi: 10.1172/JCI42090.
    1. Azuma A, et al. Interferon-β inhibits bleomycin-induced lung fibrosis by decreasing transforming growth factor-β and thrombospondin. Am. J. Respir. Cell Mol. Biol. 2005;32:93–98. doi: 10.1165/rcmb.2003-0374OC.
    1. King TE, Jr, et al. Effect of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis (INSPIRE): a multicentre, randomised, placebo-controlled trial. Lancet. 2009;374:222–228. doi: 10.1016/S0140-6736(09)60551-1.
    1. Shen B, et al. Proteomic and metabolomic characterization of COVID-19 patient sera. Cell. 2020;182:59–72. doi: 10.1016/j.cell.2020.05.032.
    1. Bojkova D, et al. Proteomics of SARS-CoV-2-infected host cells reveals therapy targets. Nature. 2020;583:469–472. doi: 10.1038/s41586-020-2332-7.
    1. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30. doi: 10.1093/nar/28.1.27.
    1. Antonio GE, et al. Thin-section CT in patients with severe acute respiratory syndrome following hospital discharge: preliminary experience. Radiology. 2003;228:810–815. doi: 10.1148/radiol.2283030726.
    1. Geyer PE, Holdt LM, Teupser D, Mann M. Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol. 2017;13:942. doi: 10.15252/msb.20156297.
    1. Bensadoun ES, Burke AK, Hogg JC, Roberts CR. Proteoglycan deposition in pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 1996;154:1819–1828. doi: 10.1164/ajrccm.154.6.8970376.
    1. Li Y, et al. Hyaluronan synthase 2 regulates fibroblast senescence in pulmonary fibrosis. Matrix Biol. 2016;55:35–48. doi: 10.1016/j.matbio.2016.03.004.
    1. Liang J, et al. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. Nat. Med. 2016;22:1285–1293. doi: 10.1038/nm.4192.
    1. Cavallaro U, Dejana E. Adhesion molecule signalling: not always a sticky business. Nat. Rev. Mol. Cell Biol. 2011;12:189–197. doi: 10.1038/nrm3068.
    1. Harjunpää H, Llort AM, Guenther C, Fagerholm SC. Cell adhesion molecules and their roles and regulation in the immune and tumor microenvironment. Front. Immunol. 2019;10:1078. doi: 10.3389/fimmu.2019.01078.
    1. Marelli-Berg FM, Clement M, Mauro C, Caligiuri G. An immunologist's guide to CD31 function in T-cells. J. Cell Sci. 2013;126:2343–2352. doi: 10.1242/jcs.124099.
    1. Privratsky JR, Newman DK, Newman PJ. PECAM-1: conflicts of interest in inflammation. Life Sci. 2010;87:69–82. doi: 10.1016/j.lfs.2010.06.001.
    1. Villar J, Zhang H, Slutsky AS. Lung repair and regeneration in ARDS: role of PECAM1 and wnt signaling. Chest. 2019;155:587–594. doi: 10.1016/j.chest.2018.10.022.
    1. Watson WH, Ritzenthaler JD, Roman J. Lung extracellular matrix and redox regulation. Redox Biol. 2016;8:305–315. doi: 10.1016/j.redox.2016.02.005.
    1. Cox N, Pilling D, Gomer RH. Distinct Fcγ receptors mediate the effect of serum amyloid p on neutrophil adhesion and fibrocyte differentiation. J. Immunol. 2014;193:1701–1708. doi: 10.4049/jimmunol.1400281.
    1. Pilling D, Buckley CD, Salmon M, Gomer RH. Inhibition of fibrocyte differentiation by serum amyloid P. J. Immunol. 2003;171:5537–5546. doi: 10.4049/jimmunol.171.10.5537.
    1. Song JW, et al. Immunological and inflammatory profiles in mild and severe cases of COVID-19. Nat. Commun. 2020;11:3410. doi: 10.1038/s41467-020-17240-2.
    1. Bournazos S, et al. Copy number variation of FCGR3B is associated with susceptibility to idiopathic pulmonary fibrosis. Respiration. 2011;81:142–149. doi: 10.1159/000321997.
    1. Castaño AP, et al. Serum amyloid P inhibits fibrosis through Fc gamma R-dependent monocyte-macrophage regulation in vivo. Sci. Transl. Med. 2009;1:5ra3. doi: 10.1126/scitranslmed.3000111.
    1. Satoh T, et al. Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature. 2017;541:96–101. doi: 10.1038/nature20611.
    1. Oh JS, et al. NK cells lacking FcεRIγ are associated with reduced liver damage in chronic hepatitis C virus infection. Eur. J. Immunol. 2016;46:1020–1029. doi: 10.1002/eji.201546009.
    1. Asai Y, et al. Aberrant populations of circulating T follicular helper cells and regulatory B cells underlying idiopathic pulmonary fibrosis. Respir. Res. 2019;20:244. doi: 10.1186/s12931-019-1216-6.
    1. Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat. Rev. Endocrinol. 2017;13:36–49. doi: 10.1038/nrendo.2016.135.
    1. Kheirollahi V, et al. Metformin induces lipogenic differentiation in myofibroblasts to reverse lung fibrosis. Nat. Commun. 2019;10:2987. doi: 10.1038/s41467-019-10839-0.
    1. Bargagli E, et al. Metabolic dysregulation in idiopathic pulmonary fibrosis. Int. J. Mol. Sci. 2020;21:5663. doi: 10.3390/ijms21165663.
    1. Landi C, et al. A system biology study of BALF from patients affected by idiopathic pulmonary fibrosis (IPF) and healthy controls. Proteomics Clin. Appl. 2014;8:932–950. doi: 10.1002/prca.201400001.
    1. Avouac J, et al. Pan-PPAR agonist IVA337 is effective in experimental lung fibrosis and pulmonary hypertension. Ann. Rheum. Dis. 2017;76:1931–1940. doi: 10.1136/annrheumdis-2016-210821.
    1. El Agha E, et al. Two-way conversion between lipogenic and myogenic fibroblastic phenotypes marks the progression and resolution of lung fibrosis. Cell Stem Cell. 2017;20:261–273. doi: 10.1016/j.stem.2016.10.004.
    1. Jain M, et al. Leptin promotes fibroproliferative acute respiratory distress syndrome by inhibiting peroxisome proliferator-activated receptor-γ. Am. J. Respir. Crit. Care Med. 2011;183:1490–1498. doi: 10.1164/rccm.201009-1409OC.
    1. Steinritz D, Stenger B, Dietrich A, Gudermann T, Popp T. TRPs in tox: involvement of transient receptor potential-channels in chemical-induced organ toxicity-a structured review. Cells. 2018;7:98. doi: 10.3390/cells7080098.
    1. Grace MS, Baxter M, Dubuis E, Birrell MA, Belvisi MG. Transient receptor potential (TRP) channels in the airway: role in airway disease. Br. J. Pharmacol. 2014;171:2593–2607. doi: 10.1111/bph.12538.
    1. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N. Engl. J. Med. 1993;329:2002–2012. doi: 10.1056/NEJM199312303292706.
    1. Mehta S, Stewart DJ, Langleben D, Levy RD. Short-term pulmonary vasodilation with L-arginine in pulmonary hypertension. Circulation. 1995;92:1539–1545. doi: 10.1161/01.CIR.92.6.1539.
    1. Gao L, et al. Combination of L-Arginine and L-Norvaline protects against pulmonary fibrosis progression induced by bleomycin in mice. Biomed. Pharmacother. 2019;113:108768. doi: 10.1016/j.biopha.2019.108768.
    1. Ellul MA, et al. Neurological associations of COVID-19. Lancet Neurol. 2020;19:767–783. doi: 10.1016/S1474-4422(20)30221-0.
    1. China NHCO. New coronavirus pneumonia diagnosis and treatment scheme (6th edn). . Accessed 18 Feb 2020.
    1. Chung M, et al. CT imaging features of 2019 novel coronavirus (2019-nCoV) Radiology. 2020;295:202–207. doi: 10.1148/radiol.2020200230.
    1. Bernheim, A., et al. Chest CT findings in coronavirus disease-19 (COVID-19): relationship to duration of infection. Radiology295, 200463 (2020).
    1. Ma J, et al. iProX: an integrated proteome resource. Nucleic Acids Res. 2019;47:D1211–D1217. doi: 10.1093/nar/gky869.

Source: PubMed

3
Subscribe