EC Pulmonology and Respiratory Medicine

Review Article Volume 14 Issue 1 - 2025

Overview of Multi-Omics Approaches for Pulmonary Sarcoidosis

Yifan Fei1, Zhe Lei1,2,3, Yuhong Wang1,2, Macaluso Joshua3, Lisa A Maier3,4, Lingchuan Guo1,2* and Li Li3,4*

1Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China

2Institute of Clinical Pathology and Precision Medicine, Soochow University, Suzhou, Jiangsu, China

3Department of Medicine, National Jewish Health, Denver, CO, USA

4Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA

*Corresponding Author: Lingchuan Guo, Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China. E-mail: szglc@hotmail.com and Li Li, Division of Environmental and Occupational Health Sciences, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA; E-mail: lil@njhealth.org.
Received: October 28, 2024; Published:December 16, 2024



Purpose: Here, we review recent findings in the transcriptome, proteome, metabolomics, and microbiome of pulmonary sarcoidosis and highlight differentially expressed genes, specific pathways, mechanisms, microorganisms, metabolites, and targeted therapeutics in the field.

Recent Findings: The transcriptome and proteome of pulmonary sarcoidosis have been widely studied in recent years. Many differentially expressed genes and signaling pathways have been identified. Several proteins have been identified as potential molecular markers of pulmonary sarcoidosis. The microorganisms and metabolites of patients with sarcoidosis also have certain specificity. We compared pulmonary sarcoidosis with other diseases, such as idiopathic pulmonary fibrosis, tuberculosis, and chronic beryllium disease, and found some differential diagnoses. Based on the identified pathways and mechanisms, targeted therapeutic strategies have been proposed.

Summary: Many differentially expressed genes have been identified, including CBX8, CCL5, CXCL9, CXCL11, GBP1, GBP5, LINC01278, MMP12, PSMB9, STAT1, and TLE3, as well as the related enriched pathways, such as the IFN-γ, IL-1, IL-17, MHC, T-cell receptor, TNF, Th1, and Th2 signaling pathways. Proteins such as ABCG1, Apo A–I, CXCR5, MMP12, PD-1, PPARγ, and vitamin D-binding protein, together with the Fc galactosylation status of IgG4, are potential molecular markers for pulmonary sarcoidosis. Many specific microorganisms and metabolites in patients with sarcoidosis have also been found. Targeted drugs such as infliximab, nintedanib and rituximab have been proposed according to the discovered pathways and mechanisms.

 Keywords: Pulmonary Sarcoidosis; Transcriptome; Proteome; Metabolomics; Microbiome; Multi-Omics

  1. Tana C., et al. “Clinical features, histopathology and differential diagnosis of sarcoidosis”. Cells1 (2021): 59.
  2. Crouser ED., et al. “Diagnosis and Detection of Sarcoidosis. An Official American Thoracic Society Clinical Practice Guideline”. American Journal of Respiratory and Critical Care Medicine 8 (2020): e26-e51.
  3. Ozsolak F and Milos PM. “RNA sequencing: advances, challenges and opportunities”. Nature Reviews Genetics 2 (2011): 87-98.
  4. Koth LL., et al. “Defining CD4 T helper and T regulatory cell endotypes of progressive and remitting pulmonary sarcoidosis (BRITE): protocol for a US-based, multicentre, longitudinal observational bronchoscopy study”. BMJ Open11 (2021): e056841.
  5. Ascoli C., et al. “Altered transcription factor targeting is associated with differential peripheral blood mononuclear cell proportions in sarcoidosis”. Frontiers in Immunology 13 (2022): 848759.
  6. Yoshioka K., et al. “Transcriptome analysis of peripheral blood mononuclear cells in pulmonary sarcoidosis”. Frontiers in Medicine 9 (2022): 822094.
  7. Talreja J., et al. “RNA-sequencing identifies novel pathways in sarcoidosis monocytes”. Scientific Reports 1 (2017): 2720.
  8. Benn BS., et al. “Clinical and biological insights from the university of California San Francisco prospective and longitudinal cohort”. Lung 5 (2017): 553-561.
  9. Jiang Y., et al. “A transcriptomics-based meta-analysis identifies a cross-tissue signature for sarcoidosis”. Frontiers in Medicine 9 (2022): 960266.
  10. Bhargava M., et al. “The landscape of transcriptomic and proteomic studies in sarcoidosis”. ERJ Open Research 1 (2022): 00621-02021.
  11. Bauer L., et al. “Follicular helper-like T cells in the lung highlight a novel role of B cells in sarcoidosis”. American Journal of Respiratory and Critical Care Medicine12 (2021): 1403-1417.
  12. Lepzien R., et al. “Pulmonary and blood dendritic cells from sarcoidosis patients more potently induce IFNγ-producing Th1 cells compared with monocytes”. Journal of Leukocyte Biology 4 (2022): 857-866.
  13. Vukmirovic M., et al. “Transcriptomics of bronchoalveolar lavage cells identifies new molecular endotypes of sarcoidosis”. European Respiratory Journal 6 (2021): 2002950.
  14. Ed C., et al. “Gene expression profiling identifies MMP-12 and ADAMDEC1 as potential pathogenic mediators of pulmonary sarcoidosis”. American Journal of Respiratory and Critical Care Medicine10 (2009): 929-938.
  15. Lepzien R., et al. “Monocytes in sarcoidosis are potent tumour necrosis factor producers and predict disease outcome”. European Respiratory Journal 1 (2021): 2003468.
  16. Talreja J., et al. “HIF-1α regulates IL-1β and IL-17 in sarcoidosis”. eLife 8 (2019): e44519.
  17. Locke LW., et al. “IL-13-regulated macrophage polarization during granuloma formation in an in vitro human sarcoidosis model”. American Journal of Respiratory Cell and Molecular Biology 1 (2019): 84-95.
  18. Mohan A., et al. “Transcriptional survey of alveolar macrophages in a murine model of chronic granulomatous inflammation reveals common themes with human sarcoidosis”. American Journal of Physiology-Lung Cellular and Molecular Physiology 4 (2018): L617-L625.
  19. Kachamakova-Trojanowska N., et al. “Molecular profiling of regulatory T cells in pulmonary sarcoidosis”. Journal of Autoimmunity 94 (2018): 56-69.v
  20. Krausgruber T., et al. “Single-cell and spatial transcriptomics reveal aberrant lymphoid developmental programs driving granuloma formation”. Immunity2 (2023): 289-306.e7.
  21. Bhargava M., et al. “Novel protein pathways in development and progression of pulmonary sarcoidosis”. Scientific Reports 1 (2020): 13282.
  22. Martinez-Bravo MJ., et al. “Pulmonary sarcoidosis is associated with exosomal vitamin D-binding protein and inflammatory molecules”. Journal of Allergy and Clinical Immunology 4 (2017): 1186-1194.v
  23. Nukui Y., et al. “Identification of apolipoprotein A-I in BALF as a biomarker of sarcoidosis”. Sarcoidosis, Vasculitis and Diffuse Lung Diseases 1 (2018): 5-15.
  24. Kim KD., et al. “Apolipoprotein A-I induces IL-10 and PGE2 production in human monocytes and inhibits dendritic cell differentiation and maturation”. Biochemical and Biophysical Research Communications 2 (2005): 1126-1136.
  25. Barna BP., et al. “Studies in a murine granuloma model of instilled carbon nanotubes: relevance to sarcoidosis”. International Journal of Molecular Sciences 7 (2021): 3705.
  26. Isshiki T., et al. “Plasma matrix metalloproteinase 7, CC-chemokine ligand 18, and periostin as markers for pulmonary sarcoidosis”. Respiratory Investigation 6 (2020): 479-487.
  27. Bargagli E., et al. “The effect of cigarette smoking on bronchoalveolar lavage protein profiles from patients with different interstitial lung diseases”. Panminerva Medica 2 (2020): 109-115.
  28. Heyder T., et al. “Altered Fc galactosylation in IgG4 is a potential serum marker for chronic lung disease”. ERJ Open Research 3 (2018): 00033-02018.
  29. Geamanu A., et al. “Metabolomics connects aberrant bioenergetic, transmethylation, and gut microbiota in sarcoidosis”. Metabolomics 12 (2016): 35.
  30. Suzuki T., et al. “Associations of serum long-chain fatty acids with multiple organ involvement in patients with sarcoidosis”. BMC Pulmonary Medicine 1 (2022): 290.
  31. Duchemann B., et al. “Nuclear magnetic resonance spectroscopic analysis of salivary metabolome in sarcoidosis”. Sarcoidosis, Vasculitis and Diffuse Lung Diseases 1 (2016): 10-16.
  32. Mirsaeidi M., et al. “Plasma metabolomic profile in fibrosing pulmonary sarcoidosis”. Sarcoidosis, Vasculitis and Diffuse Lung Diseases 1 (2016): 29-38.
  33. Banoei MM., et al. “Metabolomic and metallomic profile differences between Veterans and Civilians with Pulmonary Sarcoidosis”. Scientific Reports 1 (2019): 19584.
  34. Gupta S., et al. “Comparative analysis of the alveolar microbiome in COPD, ECOPD, Sarcoidosis, and ILD patients to identify respiratory illnesses specific microbial signatures”. Scientific Reports 1 (2021): 3963.
  35. D’Argenio V., et al. “A common microbial signature is present in the lower airways of interstitial lung diseases including sarcoidosis”. Sarcoidosis, Vasculitis and Diffuse Lung Diseases 4 (2018): 354-362.
  36. Zimmermann A., et al. “Atopobium and Fusobacterium as novel candidates for sarcoidosis-associated microbiota”. European Respiratory Journal 6 (2017): 1600746.
  37. Becker A., et al. “The composition of the pulmonary microbiota in sarcoidosis - an observational study”. Respiratory Research 1 (2019): 46.
  38. Garzoni C., et al. “Microbial communities in the respiratory tract of patients with interstitial lung disease”. Thorax12 (2013): 1150-1156.
  39. Knudsen KS., et al. “The lower airways microbiota and antimicrobial peptides indicate dysbiosis in sarcoidosis”. Microbiome1 (2022): 175.
  40. Shi H., et al. “The associations between gut microbiota and chronic respiratory diseases: a Mendelian randomization study”. Frontiers in Microbiology 14 (2023): 1200937.
  41. Chioma OS., et al. “Low gut microbial diversity augments estrogen-driven pulmonary fibrosis in female-predominant interstitial lung disease”. Cells5 (2023): 766.
  42. Konigsberg IR., et al. “Multi-omic signatures of sarcoidosis and progression in bronchoalveolar lavage cells”. bioRxiv (2023): 2023.01.26.525601.
  43. Hočevar K., et al. “Sarcoidosis related novel candidate genes identified by multi-omics integrative analyses”. OMICS: A Journal of Integrative Biology 5 (2018): 322-331.
  44. Kamp JC., et al. “Comparative analysis of gene expression in fibroblastic foci in patients with idiopathic pulmonary fibrosis and pulmonary sarcoidosis”. Cells4 (2022): 664.
  45. Majewski S., et al. “Proteomic profiling of peripheral blood and bronchoalveolar lavage fluid in interstitial lung diseases: an explorative study”. ERJ Open Research 1 (2021): 00489-02020.
  46. Bennett D., et al. “Elevated level of Galectin-1 in bronchoalveolar lavage of patients with idiopathic pulmonary fibrosis”. Respiratory Physiology and Neurobiology 273 (2020): 103323.
  47. Paplińska-Goryca M., et al. “mRNA expression profile of bronchoalveolar lavage fluid cells from patients with idiopathic pulmonary fibrosis and sarcoidosis”. European Journal of Clinical Investigation 9 (2019): e13153.
  48. Prasse A., et al. “BAL cell gene expression is indicative of outcome and airway basal cell involvement in idiopathic pulmonary fibrosis”. American Journal of Respiratory and Critical Care Medicine 5 (2019): 622-630.
  49. Gangwar I., et al. “Detecting the molecular system signatures of idiopathic pulmonary fibrosis through integrated genomic analysis”. Scientific Reports 1 (2017): 1554.v
  50. Lee JU., et al. “Gene profile of fibroblasts identify relation of CCL8 with idiopathic pulmonary fibrosis”. Respiratory Research 1 (2017): 3.
  51. Reichmann MT., et al. “Integrated transcriptomic analysis of human tuberculosis granulomas and a biomimetic model identifies therapeutic targets”. Journal of Clinical Investigation 15 (2021): e148136.
  52. Casanova NG., et al. “Differential transcriptomics in sarcoidosis lung and lymph node granulomas with comparisons to pathogen-specific granulomas”. Respiratory Research 1 (2020): 321.
  53. Chai Q., et al. “Lung gene expression signatures suggest pathogenic links and molecular markers for pulmonary tuberculosis, adenocarcinoma and sarcoidosis”. Communications Biology 1 (2020): 604.
  54. Zhao M., et al. “Identification of biomarkers for sarcoidosis and tuberculosis of the lung using systematic and integrated analysis”. Medical Science Monitor 26 (2020): e925438.
  55. Liao SY., et al. “Single-cell RNA sequencing identifies macrophage transcriptional heterogeneities in granulomatous diseases”. European Respiratory Journal 6 (2021): 2003794.
  56. Lin NW., et al. “Genomic biomarkers in chronic beryllium disease and sarcoidosis”. Respiratory Medicine 187 (2021): 106390.
  57. Li L., et al. “Beryllium-induced lung disease exhibits expression profiles similar to sarcoidosis”. European Respiratory Journal6 (2016): 1797-1808.
  58. Tanaka H., et al. “Cathepsin S, a new serum biomarker of sarcoidosis discovered by transcriptome analysis of alveolar macrophages”. Sarcoidosis, Vasculitis and Diffuse Lung Diseases 2 (2019): 141-147.
  59. Komatsu M., et al. “The utility of serum C-C chemokine ligand 1 in sarcoidosis: A comparison to IgG4-related disease”. Cytokine 133 (2020): 155123.
  60. Yamamoto H., et al. “Comparison of the chemokine profiles in the bronchoalveolar lavage fluid between IgG4-related respiratory disease and sarcoidosis: CC-chemokine ligand 1 might be involved in the pathogenesis of sarcoidosis”. Cytokine 120 (2019): 125-129.
  61. Meng K., et al. “Serum amyloid A/anti-CCL20 induced the rebalance of Th17/regulatory T cells in SodA-induced sarcoidosis”. International Immunopharmacology 109 (2022): 108784.
  62. Wahlund CJE., et al. “Sarcoidosis exosomes stimulate monocytes to produce pro-inflammatory cytokines and CCL2”. Scientific Reports 1 (2020): 15328.
  63. Huppertz C., et al. “The NLRP3 inflammasome pathway is activated in sarcoidosis and involved in granuloma formation”. European Respiratory Journal 3 (2020): 1900119.
  64. Lo CY., et al. “Increased interleukin-17 and glucocorticoid receptor-β expression in interstitial lung diseases and corticosteroid insensitivity”. Frontiers in Immunology 13 (2022): 905727.
  65. C K., et al. “Sarcoidosis manifesting during treatment with secukinumab for psoriatic arthritis”. BMJ Case Reports2 (2021): bcr-2020-240615.
  66. Song J., et al. “IL-17A can promote Propionibacterium acnes-induced sarcoidosis-like granulomatosis in mice”. Frontiers in Immunology 10 (2019): 1923.
  67. W D., et al. “Inhibition of type 1 immunity with tofacitinib is associated with marked improvement in longstanding sarcoidosis”. Nature Communications 1 (2022): 3140.
  68. Zhang H., et al. “The role of diverse immune cells in sarcoidosis”. Frontiers in Immunology 12 (2021): 788502.
  69. A W., et al. “The promise of JAK inhibitors for treatment of sarcoidosis and other inflammatory disorders with macrophage activation: a review of the literature”. Yale Journal of Biology and Medicine 1 (2020): 187-195.
  70. Richmond BW., et al. “Sarcoidosis Th17 cells are ESAT-6 antigen specific but demonstrate reduced IFN-γ expression”. Journal of Clinical Immunology2 (2013): 446-455.
  71. Rezaee M., et al. “Role of anti-tumor necrosis factor-alpha agents in treatment of sarcoidosis: A meta-analysis”. European Journal of Internal Medicine 109 (2022): 42-49.
  72. Singh K., et al. “Novel Th17 lymphocyte populations, Th17.1 and PD1+Th17, are increased in Takayasu arteritis, and both Th17 and Th17.1 sub-populations associate with active disease”. Journal of Inflammation Research 15 (2022): 1521-1541.
  73. Hamada E., et al. “Pulmonary sarcoidosis with a cavitary lesion in the lung caused by a TNF-α inhibitor: A case report”. Respirology Case Reports 12 (2022): e01065.
  74. Malur A., et al. “Peroxisome proliferator-activated receptor-γ deficiency exacerbates fibrotic response to Mycobacteria peptide in murine sarcoidosis model”. American Journal of Respiratory Cell and Molecular Biology 2 (2019): 198-208.
  75. Mohan A., et al. “Matrix metalloproteinase-12 is required for granuloma progression”. Frontiers in Immunology 11 (2020): 553949.
  76. Ogburn D., et al. “The M2a macrophage phenotype accompanies pulmonary granuloma resolution in Mmp12 knock-out mice instilled with multiwall carbon nanotubes”. International Journal of Molecular Sciences 20 (2021): 11019.v
  77. West SG. “Current management of sarcoidosis I: pulmonary, cardiac, and neurologic manifestations”. Current Opinion in Rheumatology 3 (2018): 243-248.
  78. Talreja J., et al. “Derangement of metabolic and lysosomal gene profiles in response to dexamethasone treatment in sarcoidosis”. Frontiers in Immunology 11 (2020): 779.
  79. Arakelyan A., et al. “Transcriptome-guided drug repositioning”. Pharmaceutics12 (2019): 677.
  80. Comes A., et al. “Novel insights in fibrotic pulmonary sarcoidosis”. Current Opinion in Pulmonary Medicine 5 (2022): 478-484.

Lingchuan Guo and Li Li., et al. "Overview of Multi-Omics Approaches for Pulmonary Sarcoidosis". EC Pulmonology and Respiratory Medicine  14.1 (2025): 01-16.