DOI QR코드

DOI QR Code

Increased Inflammatory Responses in Patients With Active Disseminated Non-Tuberculous Mycobacterial Infection and High Anti-Interferon-Gamma Autoantibodies

  • Pattaraporn Srisai (Department of Microbiology, Faculty of Medicine, Khon Kaen University) ;
  • Chanchai Hongsa (Department of Microbiology, Faculty of Medicine, Khon Kaen University) ;
  • Yothin Hinwan (Department of Microbiology, Faculty of Medicine, Khon Kaen University) ;
  • Varis Manbenmad (Department of Microbiology, Faculty of Medicine, Khon Kaen University) ;
  • Ploenchan Chetchotisakd (Department of Medicine, Faculty of Medicine Khon Kaen University) ;
  • Siriluck Anunnatsiri (Department of Medicine, Faculty of Medicine Khon Kaen University) ;
  • Kiatichai Faksri (Department of Microbiology, Faculty of Medicine, Khon Kaen University) ;
  • Todsapol Techo (Department of Biology, Faculty of Science, Khon Kaen University) ;
  • Kanin Salao (Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore) ;
  • Steven W. Edwards (Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool) ;
  • Arnone Nithichanon (Department of Microbiology, Faculty of Medicine, Khon Kaen University)
  • Received : 2024.07.16
  • Accepted : 2024.09.06
  • Published : 2024.10.31

Abstract

Adult-onset immunodeficiency (AOID) is associated with the presence of anti-IFN-γ autoantibodies (auAbs). In disseminated nontuberculous mycobacterial (dNTM) infection with AOID, neutralization of IFN-γ by auAb may play a role in disease susceptibility, but other molecular mechanisms are likely to contribute. In this study, dNTM patients, including inactive, active but non-progressive and active, progressive cases were enrolled to measure plasma anti-IFN-γ auAb by ELISA and underwent whole-blood RNA sequencing. Healthy control individuals were also enrolled. Plasma IL-8 was then quantified to confirm transcriptomic analysis. Results revealed that anti-IFN-γ auAb titers were significantly increased in patients with active stage of disease. Gene expression could separate patients with active infection from individuals with no signs of infection (inactive patients and healthy controls). In active cases, there was over-expression of inflammatory pathways and under-expression of type-2 immunity pathways. Interestingly, increased levels of plasma IL-8 (p=0.0167) not only confirmed gene expression results but also correlated with the presence of neutrophilic dermatitis (p=0.0244). In conclusion, our findings highlight the value of anti-IFN-γ auAb titers for predicting disease reactivity and first propose IL-8 as a promising mediator to be further explored, given its correlation with skin reactive disease, a hallmark of active dNTM infection.

Keywords

Acknowledgement

This work was supported by the National Research Council of Thailand (NRCT) under contract no. N42A650205. We acknowledge the medical staffs at Srinagarind Hospital, Khon Kaen University, Thailand, for assisting and reviewing patient records. We would like to acknowledge Prof.David Blair for editing the manuscript via Publication Clinic KKU, Thailand.

References

  1. Chetchotisakd P, Kiertiburanakul S, Mootsikapun P, Assanasen S, Chaiwarith R, Anunnatsiri S. Disseminated nontuberculous mycobacterial infection in patients who are not infected with HIV in Thailand. Clin Infect Dis 2007;45:421-427.
  2. Kim RD, Greenberg DE, Ehrmantraut ME, Guide SV, Ding L, Shea Y, Brown MR, Chernick M, Steagall WK, Glasgow CG, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med 2008;178:1066-1074.
  3. Henkle E, Winthrop KL. Nontuberculous mycobacteria infections in immunosuppressed hosts. Clin Chest Med 2015;36:91-99.
  4. Patel SY, Ding L, Brown MR, Lantz L, Gay T, Cohen S, Martyak LA, Kubak B, Holland SM. Anti-IFN-gamma autoantibodies in disseminated nontuberculous mycobacterial infections. J Immunol 2005;175:4769-4776.
  5. Wongkulab P, Wipasa J, Chaiwarith R, Supparatpinyo K. Autoantibody to interferon-gamma associated with adult-onset immunodeficiency in non-HIV individuals in Northern Thailand. PLoS One 2013;8:e76371.
  6. Chi CY, Chu CC, Liu JP, Lin CH, Ho MW, Lo WJ, Lin PC, Chen HJ, Chou CH, Feng JY, et al. Anti-IFN-γ autoantibodies in adults with disseminated nontuberculous mycobacterial infections are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02 and the reactivation of latent varicella-zoster virus infection. Blood 2013;121:1357-1366.
  7. Kampmann B, Hemingway C, Stephens A, Davidson R, Goodsall A, Anderson S, Nicol M, Scholvinck E, Relman D, Waddell S, et al. Acquired predisposition to mycobacterial disease due to autoantibodies to IFN-gamma. J Clin Invest 2005;115:2480-2488.
  8. Wu UI, Holland SM. Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis 2015;15:968-980.
  9. Krisnawati DI, Liu YC, Lee YJ, Wang YT, Chen CL, Tseng PC, Lin CF. Functional neutralization of anti-IFN-γ autoantibody in patients with nontuberculous mycobacteria infection. Sci Rep 2019;9:5682.
  10. Nithichanon A, Chetchotisakd P, Matsumura T, Takahashi Y, Ato M, Sakagami T, Lertmemongkolchai G. Diagnosis of NTM active infection in lymphadenopathy patients with anti-interferon-gamma auto-antibody using inhibitory ELISA vs. indirect ELISA. Sci Rep 2020;10:8968.
  11. Chetchotisakd P, Anunnatsiri S, Nithichanon A, Lertmemongkolchai G. Cryptococcosis in antiinterferon-gamma autoantibody-positive patients: a different clinical manifestation from HIV-infected patients. Jpn J Infect Dis 2016;70:69-74.
  12. Aoki A, Sakagami T, Yoshizawa K, Shima K, Toyama M, Tanabe Y, Moro H, Aoki N, Watanabe S, Koya T, et al. Clinical significance of interferon-gamma neutralizing autoantibodies against disseminated nontuberculous mycobacterial disease. Clin Infect Dis 2018;66:1239-1245. 
  13. Chetchotisakd P, Anunnatsiri S, Nanagara R, Nithichanon A, Lertmemongkolchai G. Intravenous cyclophosphamide therapy for anti-IFN-gamma autoantibody-associated Mycobacterium abscessus infection. J Immunol Res 2018;2018:6473629.
  14. Chaussabel D. Assessment of immune status using blood transcriptomics and potential implications for global health. Semin Immunol 2015;27:58-66.
  15. Prebensen C, Lefol Y, Myhre PL, Luders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023;13:10368.
  16. Akthar M, Nair N, Carter LM, Vital EM, Sutton E, McHugh N, ; British Isles Lupus Assessment Group Biologics Register (BILAG BR) ConsortiumMASTERPLANS Consortium, Bruce IN, Reynolds JA. Deconvolution of whole blood transcriptomics identifies changes in immune cell composition in patients with systemic lupus erythematosus (SLE) treated with mycophenolate mofetil. Arthritis Res Ther 2023;25:111.
  17. Gandhi KS, McKay FC, Cox M, Riveros C, Armstrong N, Heard RN, Vucic S, Williams DW, Stankovich J, Brown M, et al. The multiple sclerosis whole blood mRNA transcriptome and genetic associations indicate dysregulation of specific T cell pathways in pathogenesis. Hum Mol Genet 2010;19:2134-2143.
  18. Parnell GP, McLean AS, Booth DR, Armstrong NJ, Nalos M, Huang SJ, Manak J, Tang W, Tam OY, Chan S, et al. A distinct influenza infection signature in the blood transcriptome of patients with severe community-acquired pneumonia. Crit Care 2012;16:R157.
  19. Panousis NI, Bertsias GK, Ongen H, Gergianaki I, Tektonidou MG, Trachana M, Romano-Palumbo L, Bielser D, Howald C, Pamfil C, et al. Combined genetic and transcriptome analysis of patients with SLE: distinct, targetable signatures for susceptibility and severity. Ann Rheum Dis 2019;78:1079-1089.
  20. Kiratikanon S, Phinyo P, Rujiwetpongstorn R, Patumanond J, Tungphaisal V, Mahanupab P, Chaiwarith R, Tovanabutra N, Chiewchanvit S, Chuamanochan M. Adult-onset immunodeficiency due to anti-interferon-gamma autoantibody-associated Sweet syndrome: a distinctive entity. J Dermatol 2022;49:133-141.
  21. Nithichanon A, Samer W, Chetchotisakd P, Kewcharoenwong C, Ato M, Lertmemongkolchai G. Evaluation of plasma anti-GPL-core IgA and IgG for diagnosis of disseminated non-tuberculous mycobacteria infection. PLoS One 2020;15:e0242598.
  22. Manbenmad V, So-Ngern A, Chetchotisakd P, Faksri K, Ato M, Nithichanon A, Lertmemongkolchai G. Evaluating anti-GPL-core IgA as a diagnostic tool for non-tuberculous mycobacterial infections in Thai patients with high antibody background. Sci Rep 2023;13:18883.
  23. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114-2120.
  24. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol 2019;37:907-915.
  25. Pott F, Postmus D, Brown RJ, Wyler E, Neumann E, Landthaler M, Goffinet C. Single-cell analysis of arthritogenic alphavirus-infected human synovial fibroblasts links low abundance of viral RNA to induction of innate immunity and arthralgia-associated gene expression. Emerg Microbes Infect 2021;10:2151-2168.
  26. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014;30:923-930.
  27. Anders S, Pyl PT, Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 2015;31:166-169.
  28. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
  29. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 2016;32:2847-2849.
  30. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 2012;16:284-287.
  31. Yu G, He QY. ReactomePA: an R/Bioconductor package for Reactome pathway analysis and visualization. Mol Biosyst 2016;12:477-479.
  32. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010;26:139-140.
  33. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, Khodadoust MS, Esfahani MS, Luca BA, Steiner D, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol 2019;37:773-782.
  34. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M, Alizadeh AA. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 2015;12:453-457.
  35. Baggiolini M, Walz A, Kunkel SL. Neutrophil-activating peptide-1/interleukin 8, a novel cytokine that activates neutrophils. J Clin Invest 1989;84:1045-1049.
  36. Chieosilapatham P, Daroontum T, Suwansirikul S, Chaiwarith R, Phinyo P, Chaowattanapanit S, Choonhakarn C, Kiratikanon S, Rujiwetpongstorn R, Tovanabutra N, et al. Comparative immunohistochemical analysis of inflammatory cytokines in distinct subtypes of Sweet syndrome. Front Immunol 2024;15:1355681.
  37. Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, Walsh AM, Baxi V, Pandya D, Baradet T, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med 2020;26:688-692.
  38. Kaiser R, Leunig A, Pekayvaz K, Popp O, Joppich M, Polewka V, Escaig R, Anjum A, Hoffknecht ML, Gold C, et al. Self-sustaining IL-8 loops drive a prothrombotic neutrophil phenotype in severe COVID-19. JCI Insight 2021;6:e150862.
  39. Kraft R, Herndon DN, Finnerty CC, Cox RA, Song J, Jeschke MG. Predictive value of IL-8 for sepsis and severe infections after burn injury: a clinical study. Shock 2015;43:222-227.
  40. Marzano AV, Cugno M, Trevisan V, Fanoni D, Venegoni L, Berti E, Crosti C. Role of inflammatory cells, cytokines and matrix metalloproteinases in neutrophil-mediated skin diseases. Clin Exp Immunol 2010;162:100-107.
  41. Marzano AV, Fanoni D, Antiga E, Quaglino P, Caproni M, Crosti C, Meroni PL, Cugno M. Expression of cytokines, chemokines and other effector molecules in two prototypic autoinflammatory skin diseases, pyoderma gangrenosum and Sweet's syndrome. Clin Exp Immunol 2014;178:48-56.
  42. Asea A, Kraeft SK, Kurt-Jones EA, Stevenson MA, Chen LB, Finberg RW, Koo GC, Calderwood SK. HSP70 stimulates cytokine production through a CD14-dependant pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med 2000;6:435-442.
  43. Vabulas RM, Ahmad-Nejad P, Ghose S, Kirschning CJ, Issels RD, Wagner H. HSP70 as endogenous stimulus of the Toll/interleukin-1 receptor signal pathway. J Biol Chem 2002;277:15107-15112.
  44. Hilger D, Masureel M, Kobilka BK. Structure and dynamics of GPCR signaling complexes. Nat Struct Mol Biol 2018;25:4-12.
  45. Cowman SA, Jacob J, Hansell DM, Kelleher P, Wilson R, Cookson WO, Moffatt MF, Loebinger MR. Whole-blood gene expression in pulmonary nontuberculous mycobacterial infection. Am J Respir Cell Mol Biol 2018;58:510-518.
  46. Xu C, Lu Z, Luo Y, Liu Y, Cao Z, Shen S, Li H, Liu J, Chen K, Chen Z, et al. Targeting of NLRP3 inflammasome with gene editing for the amelioration of inflammatory diseases. Nat Commun 2018;9:4092.
  47. Chawansuntati K, Rattanathammethee K, Wipasa J. Minireview: Insights into anti-interferon-γ autoantibodies. Exp Biol Med (Maywood) 2021;246:790-795.
  48. Lin CH, Chi CY, Shih HP, Ding JY, Lo CC, Wang SY, Kuo CY, Yeh CF, Tu KH, Liu SH, et al. Identification of a major epitope by anti-interferon-γ autoantibodies in patients with mycobacterial disease. Nat Med 2016;22:994-1001.
  49. Saha B, Jyothi Prasanna S, Chandrasekar B, Nandi D. Gene modulation and immunoregulatory roles of interferon gamma. Cytokine 2010;50:1-14.
  50. Chen L, Chi H, Teng J, Meng J, Zhang H, Su Y, Liu H, Ye J, Shi H, Hu Q, et al. Neutralizing anti-IFN-γ IgG was increased in patients with systemic lupus erythematosus and associated with susceptibility to infection. Clin Rheumatol 2024;43:189-198.
  51. Lee WI, Fang YF, Huang JL, You HL, Hsieh MY, Huang WT, Liang CJ, Kang CC, Wu TS. Distinct lymphocyte immunophenotyping and quantitative anti-interferon gamma autoantibodies in Taiwanese HIV-negative patients with non-tuberculous mycobacterial infections. J Clin Immunol 2023;43:717-727.