Introduction
Although involvement can occur in any organ, sarcoidosis is a complex disorder characterized by the presence of non-caseating granulomas primarily the lungs and lymph nodes1. Although the etiology(ies) is not known, independent investigations support infectious and non-infectious antigens in sarcoidosis pathogenesis. It has also been proposed that autoimmunity plays a crucial role in the pathogenesis of this disease. In this review, we will discuss the current knowledge regarding factors shown to initiate sarcoidosis (such as infectious and self-antigens), modify disease (checkpoint inhibitors, vaccines and antibody-mediated immunosuppression), as well as factors associated with disease (microbiome, cholesterol and environment). We will emphasize the cellular and molecular mechanisms that lead to granuloma formation and tissue damage.
IA. Infectious antigens associated with sarcoidosis pathogenesis
Mycobacterial antigens
One of the earliest descriptions of sarcoidosis (as a skin condition) by the French dermatologist Ernest Henri Besnier reveal the significant clinical overlap with tuberculosis. Besneir reported cutaneous lesion of “lupus pernio de la face-synovites fongueuses (scrofulo-tuberculeuses) symmetriques des extremities superieures.”2 There remains striking clinical, histologic and pathogenic overlap between sarcoidosis and tuberculosis. Molecular analysis of sarcoidosis granulomas from American patients reveals the presence of novel mycobacterial sequences among mycobacteria that cause tuberculosis-like syndrome3; mass spectral analysis of sarcoidosis diagnostic specimens reveals mycobacterial virulence factors, such as early secreted antigenic target, 6 kDa (ESAT-6)4. MTB antigens have been detected in the granulomas of Iranian sarcoidosis patients5. Antibody immune responses to mycobacterial virulence factors have been reported6. In a complementary fashion, adaptive T cell immune responses in sarcoidosis local and systemic environments reveal Th1 and Th17 responses to mycobacterial virulence factors, such as ESAT-6, superoxide dismutase A7, antigen 85A and katG8–10, as well as heat shock protein11. In need, administration of superoxide dismutase A to wildtype (WT) mice recapitulates sarcoidosis histologically and immunologically12–14. Interestingly, a Phase I investigation of antimycobacterial therapy of concomitant levaquin, ethambutol, azithromycin and rifampin reduced size and granuloma burden in cutaneous sarcoidosis lesions15; however, these results have not been confirmed in a Phase II investigation. Administration of this same regimen to pulmonary sarcoidosis did reduce immune responses to mycobacterial virulence factors but did not result in improvement in sarcoidosis forced vital capacity (FVC)16. This is lack of improvement in lung function is akin to findings seen after administrating antimycobacterial therapy to tuberculosis patients17 or those with nontuberculosis mycobacterial (NTM) infection18. Because antigens such as ESAT-6 do not persist but rather are actively secreted through the ESX-1 secretion system19, a reduction in immunity against mycobacterial virulence factors following antimycobacterial therapy correlates well with bacterial clearance20–22.
Proprionibacteria
The agent formerly known as Propionibacterium acnes, has been renamed, Cutibacterium acnes. Similarly, Cutibacterium acnes (CA) DNA has been found in areas of granulomatous and nongranulomatous involvement of sarcoidosis specimens4,23, and antibodies against CA have been detected in the sera of these patients24. Using the Clone Library Method detected Streptococcus gordonii and Cutibacterium acnes in sarcoidosis lymph nodes25. CA DNA or protein has been detected in ocular, cutaneous and pulmonary sarcoidosis specimens. Administration of heat-killed CA to wild-type mice resulted in the granulomatous inflammation and lung fibrosis that was ameliorated with antibody against IL-17A26. Also, administration of antimicrobial therapy such as azithromycin, which targets mycobacteria and cutibacteria, has been shown to resolve sarcoidosis granulomas15.
Viruses
Several viruses have been investigated as potential triggers for sarcoidosis, including human herpesvirus 6 (HHV-6)27, and lymphotrophic viruss such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV)28. The role of these viruses in the development of sarcoidosis remains controversial. The appearance of SARS-CoV-2 led many clinicians to assess for increased morbidity and mortality among sarcoidosis patients. It was determined that age at diagnosis of SARS-Cov-2 infection and comorbidities, such as diabetes and severe pulmonary disease, were associated with increased risk for hospitalization29, and that sarcoidosis patients could be safely vaccinated, even if immunosuppressed30.
Fungus
There have been numerous case reports of pulmonary sarcoidosis complicated by known fungal pathogens such as cryptococcus31, mucormycosis32 and coccidiomycosis33, but no convincing reports support that they contribute to sarcoidosis granuloma formation. A potential role for Aspergillus spp was reported by another group who found enrichment of a peptide from Aspergillus nidulans in the bronchoalveolar lavage (BAL) fluid of nine patients with Lofgren’s syndrome, an acute form of sarcoidosis. These patients also showed an increase in A. nidulans-specific IgG in their serum, compared to three controls34. Also, fungal cell wall antigens in organic dust have been demonstrated to modulate immunity in sarcoidosis peripheral blood mononuclear cells (PBMC)35. These findings support a role for fungal antigens as modulators of sarcoidosis pathogenesis.
IB. Noninfectious agents associated with sarcoidosis pathogenesis
A review of sarcoidosis innate and adaptive immunity is detailed in this journal. As cited above, many labs have reported local and systemic immune responses against microbial antigens, particularly pathogenic mycobacteria. In terms of innate immunity, potential non-mycobacterial-associated pathogen-associated molecular patterns (PAMP) include lipopolysaccharide (LPS) from the outer membrane of Gram-negative bacteria, peptidoglycan, lipoteichoic acid, bacterial DNA, viral DNA/RNA, chitin, flagellin, leucine-rich repeats (LRR), mannans in the yeast cell wall, and microbial HSPs36.
In terms of adaptive immunity, single cell and special transcriptomics of lungs from a sarcoidosis murine model and human cutaneous sarcoidosis lesions revealed that granuloma formation followed characteristic spatial patterns and involved genes linked to immunometabolism, cytokine and chemokine signaling, and extracellular matrix remodeling. Three cell types emerged as key players in granuloma formation: metabolically reprogrammed macrophages, cytokine-producing Th17.1 cells, and fibroblasts with inflammatory and tissue-remodeling phenotypes37. Independent laboratories report Th17 and T regulatory immune responses against mycobacterial virulence factors38,39. A recent report using a sarcoidosis murine model demonstrated that rescuing T regulatory cell function against mycobacterial SodA reduced granuloma burden14. Sarcoidosis patients also have an increased number of CD163+ macrophages which promote sarcoidosis granuloma formations, correlating with the M2 phenotype and sarcoidosis disease severity40.
Many investigators and clinicians view mycobacterial infections and autoimmune disease separately; however, mycobacterial infections induce autoantibody production41–44. For example, a recent investigation reveals among pulmonary tuberculosis patients the presence of rheumatoid factor, the components of complement system and antibodies against modified citrullinated vimentin45. In addition, there were complex alterations in B cells and follicular Th cell subsets, suggesting autoimmunity in mycobacterial pathogenesis45. For the purpose of this section, we expound upon the role of immunity against self-antigens, another important contributor to sarcoidosis pathogenesis.
Self Antigens
There is a clear association between sarcoidosis pathogenesis and autoimmunity. Sarcoidosis patients are more prone to being autoantibody-positive compared to controls (10.4 vs. 3%, p = 0.031). In particular, autoantibody profiling reveals anti-mitochondrial antibody-M2, anti-Ro52, anti-Ro60, anti-SSB, anti-P0, anti-CCP, anti-β2-GP, antinuclear antibodies, anti-Sm antibody, and rheumatoid factor (RF)46. The frequent presence of associated immune-mediated disease, among which autoimmune thyroid disorders, Sjögren Syndrome and Ankylosing Spondylitis are also frequent47.
Vimentin:
One of the self-antigens that have been proposed to play a role in sarcoidosis is vimentin, a cytoskeletal protein expressed in various cells, including macrophages and fibroblasts. Vimentin has been detected in granulomas and autoantibodies in the sea of sarcoidosis patients, and its expression correlates with disease severity48 49,50. Epitopes of native vimentin are antigenic also in sarcoidosis patients with good prognosis, such as HLA-DRB1*0301+ sarcoidosis patients51. It is also noteworthy that vimentin autoantibodies are not specific for sarcoidosis as post-translationally modified and native forms of vimentin are present in many other autoimmune diseases: rheumatoid arthritis, systemic lupus erythematosus, antiphospholipid syndrome, Crohn’s disease, ankylosing spondyloarthritis and idiopathic pulmonary fibrosis51.
Heat Shock Protein 70 (HSP70):
HSP70, a stress-induced protein expressed in response to cellular damage has also been implicated as an auto-antigen. HSP70 has been detected in the granulomas of sarcoidosis patients and has been shown to activate T cells and induce the release of cytokines. Autoantibodies against HSP70 have been found in the sera of sarcoidosis patients, and their levels correlate with disease activity52,53.
Serum amyloid A (SSA):
SSA is an acute phase protein produced primarily by the liver. Serum amyloid A has been identified to be abundant in sarcoidosis tissues, and this promiscuous host protein can serve as an innate ligand to regulate experimental granulomatous inflammation54. Higher levels of SSA were found localized to macrophages and giant cells within sarcoidosis granulomas, amplifying local Th1 responses to mycobacterial antigens. It has been shown that SSA mediates NF-κB activation through Toll-like receptor-2 (TLR2) in sarcoidosis bronchoalveolar lavage (BAL) macrophages; it has been suggested that this is a likely mechanism for sarcoidosis chronic lung disease55. However the SAA is not specific for sarcoidosis but present to the same degree in other chronic fibrotic lung diseases, such as hypersensitivity pneumonitis (HP), (eosinophilic) granulomatosis with polyangiitis ((E)GPA) patients and Idiopathic Pulmonary Fibrosis (IPF)56.
II. Disease associated factors by drug-induced sarcoidosis-like reaction (DISR)
Clinical Presentation-
DISR is a systemic granulomatous reaction that is indistinguishable from sarcoidosis in terms of clinical, biological, radiological and pathological presentation. DISR induced by immunotherapy is typically associated with respiratory and skin involvement57. Dermatological symptoms may precede the discovery of pulmonary abnormalities. Conversely, clinically silent intra-thoracic lymphadenopathy may be detected in 5% of patients treated with immune checkpoint inhibitors (ICIs). Other clinical features include fever, extra-thoracic lymphadenopathy, uveitis, hypercalcemia, nervous system, hepato-splenic, muscle, and osteo-articular involvement57,58. Whatever the responsible drug, the radiological picture of DISR completely resembles sarcoidosis, with typical bilateral hilar lymphadenopathy and/or perilymphatic pulmonary micronodules, and 18F-fluorodeoxyglucose uptake in involved organs on positron emission tomography scan58. Serum angiotensin-converting enzyme levels can also be increased58. Although the list is not exhaustive, below we discuss the more common drugs associated with DISR.
Immune checkpoint inhibitors-
Increased checkpoint inhibitor expression, specifically PD-1, PD-L1 and CTLA-4, has been reported on sarcoidosis granulomas and immune cells59–61. This therapy enhances the antitumor response by blocking down regulators of T-cell immunity, specifically Programmed Cell Death 1 (PD-1) and Programmed Death Ligand 1 (PD-L1). Common ICIs are antibodies targeting PD-1 (Nivolumab, Pembrolizumab), the PD-L1 (Atezolizumab, Avelumab, Durvalumab), or Cytotoxic T-lymphocyte Antigen 4 (CTLA-4) (Ipilimumab). The mean time from immunotherapy initiation to DISR onset was 7.1+ 9 months. In a series of 32 cancer patients with preexisting sarcoidosis who received immune checkpoint inhibition (ICI), only one case with a 20-year remote history had a symptomatic exacerbation requiring systemic corticosteroids62. Another retrospective study of ICI-treated patients without a prior diagnosis of sarcoidosis, DISR occurred in approximately 3.7% of patients treated with either anti-CTLA-4 or anti-PD-1 antibody and 6.3% of patients treated with a combination of both. Melanoma patients who developed de novo sarcoidosis with anti-PD-1 therapy had more Th17.1 cells in blood at baseline (pre-anti-PD-1 therapy) compared with melanoma patients who did not, suggesting that like classical sarcoidosis, Th17.1 cells might play an important role in de novo sarcoidosis-ir adverse events63,64. The mean interval from the initiation of immunotherapy to development of DISR was 5.5 months (range 2.3-13.5 months). Mean interval from radiological detection of DISR to imaging evidence of resolution was 5.8 months (range 1.6-18.3 months). Three patients out of 81 (3.7%), 11/297 (3.7%), and 5/79 (6.3%) developed sarcoidosis-like reaction after treatment with anti-CTLA-4 antibody, anti-PD-1 antibody, and a combination of both, respectively. Most patients with DISR were asymptomatic and did not require systemic therapy65,66. Further studies to assess if DISR T cells recognized mycobacteria, propionibacteria or self-antigens is warranted to further delineate if these Th17 cells are antigen-specific.
Antiretroviral therapy-
when HIV-positive patients are treated with combination antiretroviral therapy (cART) and CD4+Tcell counts rise to > 150–200 cells/microliter, a DISR indistinguishable from sarcoidosis may develop as part of immune reconstitution58,67. Additionally, cART may also exacerbate prior sarcoidosis67–69. DISRs are on average reported 9–20 months after cART initiation, although reports of Sarcoidosis occurring in HIV patients prior to cART initiation are present58,68.
Interferons-
IFN-alpha (IFNα) is produced by leukocytes, specifically by macrophages and dendritic cells. IFN-beta (IFNβ) is produced by fibroblasts. The third form of IFN is IFN-gamma (IFNγ), which is produced by T-cell lymphocytes. Lung and mediastinal lymph nodes (70%) and skin (60%) were the most commonly reported organs involved with DISRs from IFNα therapy6,45,77. IFNα-induced DISRs have been detected from 6 to 104 weeks after the beginning of therapy. While the exact mechanism associated with IFN-associated DISR, researchers suspect enhanced Th1 cell polarization, as well as induction of cytokines associated with granuloma formation70. Symptoms typically resolve with cessation of therapy or with corticosteroid use.
It is also noteworthy that some immune-mediated drugs that are used to treat sarcoidosis has also been reported to induce it, such as Tumor-necrosis factor alpha antagonists and anti-IL-6 antibody (tociclixumab).
Tumor-necrosis factor alpha antagonists-
A systematic literature review on publications with DISRs related to anti-TNFα therapy was recently conducted, excluding cases with a previous sarcoidosis diagnosis71. Over 100 DISRs were identified: 49% due to treatment with Etanercept, 25% received adalimumab and 20% of patients were treated with infliximab. Most patients were on anti-TNFα therapy for more than two years with a mean treatment duration of 25.6 months (range 1–132 months)71. Because TNFα expression is important for granuloma formation, the rare observation of sarcoidosis granuloma formation with antibody against TNFα is paradoxical. While the mechanism is unclear, neutralization of soluble TNF-α that allows activation of specific autoreactive T cells has been proposed72.
Tociclizumab-
Toziclizumab has been reported to reduce pulmonary sarcoidosis symptoms73. Paradoxically, it has also been demonstrated to induce pulmonary and/or cutaneous sarcoidosis. Both clinical phenotypes of sarcoidosis respond well to corticosteroids or cessation of the agent74.
Vaccines:
The Bacille Calmette-Guérin (BCG) vaccine, a live attenuated strain of Mycobacterium bovis, is widely used for the prevention of TB. BCG vaccination has been associated with the development of sarcoidosis granulomas in some individuals, although the incidence of this association is rare75. The pathogenesis of sarcoidosis granulomas in response to BCG vaccination is not clear, but it may be related to immune dysregulation, by shifting towards a Th1 immune response. It is important to note that sarcoidosis granulomas due to BCG vaccination is common; the vaccine remains an important tool for the prevention of tuberculosis. There is a recent investigation of the sarcoidosis incidence rate during periods of high versus low BCG vaccination among the Danish. The investigation noted that the sarcoidosis incidence rate was increased for individuals born during low BCG vaccination rates compared with individuals born during high BCG vaccination rates. The IRR of sarcoidosis for men born during low BCG vaccine uptake versus high BCG vaccine uptake was 1.22 (95% confidence interval [CI] 1.02-1.45)76. This is in contrast to a report of 54,000 English children who underwent BCG vaccination in which no efficacy against sarcoidosis was noted77,78. Other vaccines have been reported to induce sarcoidosis. For example, sarcoidosis has been reported as a possible adverse reaction to the Covid-19 Vaccine79,80,81. However, in a study monitoring autoinflammatory and autoimmune adverse events in patients that received the Covid-19 vaccine, the incidence of sarcoidosis following vaccination was rare (2 [0.3%] out of 756 patients) 82.
Adjuvants:
The adjuvants associated with autoimmune/inflammatory syndrome (ASIAs) include silicone83, hyaluronic acid84, and mineral oil85. Several sarcoidosis reactions related to ASIAs, from acute presentations such as Lofgren’s syndrome to more chronic presentations such as neurosarcoidosis, have been reported after silicone breast implantation83,86–88 with resolution of the sarcoidosis after removal of the implant89. Sarcoidosis has been reported in two cases after hip replacement90,91. Both cases were associated with systemic dissemination of prosthetic particles that were found in distant organs. These findings were suggestive of a granulomatous response to the hip prosthetic particles from wear and tear of the prosthesis. Multiple organ granulomas were found in both cases, including in the lung, liver, spleen, eye, and lymph nodes.
DISR Outcomes-
The majority of DISRs are characterized by a relatively indolent course with resolution in most cases, either spontaneously or after withdrawal of pharmacological triggers58. Specific treatment was required in 58.8% of cases, including topical corticosteroids (12.9%), systemic corticosteroids (43.5%), methotrexate (2.4%), infliximab (3.5%), and hydroxychloroquine (1.2%). After a mean follow-up of 8.3 9.9 months, DISR was stable in 12% of cases, improved in 38.7%, and resolved in 49.3%. Importantly, the decision made on immunotherapy seemed to have no impact on DISR outcomes. A table of common pharmacologic inducers of sarcoidosis, the induction period and therapeutic intervention are listed in Table 1.
Table 1.
Infectious and noninfectious drivers of sarcoidosis pathogenesis
| I. Infectious antigens | Potential Mechanism(s) | References |
|---|---|---|
| A. Mycobacteria | Secreted antigens are targets of local and systemic adaptive immune responses | 4–10 |
| B. Propionibacteria | Antigens can induce granuloma formation; antigen present in nongranulomatous regions also | 23 |
| C. Fungal antigens | Antigens augment adaptive immune response | 31–33 |
| D. Self-antigens | Present in granulomas; expression associated with disease severity | 51–56 |
| II. Disease Associated Factors | ||
| A. Drugs | ||
| 1) Checkpoint Inhibitors | Reverse PD-1, PD-L1-or CTLA-4-mediated immune dysfunction | 59, 60 |
| 2) Antiretroviral Therapy | Reverse immune dysfunction | 67–69 |
| 3) Interferons | Enhanced Th1 cell polarization; induction of cytokines associated with granuloma formation | 70 |
| 4) Immunomodulators | May permit the activation of specific autoreactive T cells. | 72 |
| B. Adjuvants | Induces foreign body granuloma formation | 83–85 |
| III. Disease Modifying Factors | ||
| A. Occupations | Associated with “Sarcoidosis clusters” | 92–95, 99 |
| B. Organic Dust | Mechanism(s) unclear | 35 |
| C. Inorganic Dust | Mechanism(s) unclear | 106 |
| D. Metals | Drive foreign-body granuloma formation | 108 |
| E. Microbiome | Dysbiotic communities drive Th17 cell differentiation | 113, 119 |
| F. Cholesterol | Aberrant lipid metabolism in sarcoidosis macrophages drives lung granuloma formation | 127 |
III. Disease modifying factors
Environment
The clinical observation of “sarcoidosis clusters” among firefighters and nurses, as well as Japanese and Swedish families suggests that environment is important92–95. Environmental factors are increasingly recognized as potential contributors to sarcoidosis development and progression. The highest prevalence of sarcoidosis in the world is in the southeastern United States among African Americans, as well as among Swedes and Danes. The highest numbers of environmental opportunistic mycobacteria are located within the brown-water swamps of the southeastern coastal United States96, as well as waters draining from boreal forest soils and peat lands in Finland97. Elucidating the role of these environmental factors and the underlying mechanisms involved could improve our understanding of sarcoidosis pathogenesis, facilitate risk assessment, and guide preventive strategies and therapeutic interventions98.
Occupational Exposures
Occupational exposure has been associated with an increased risk of developing sarcoidosis. These exposures can occur in various occupational settings, such as construction, agriculture, healthcare, and manufacturing. While the exact mechanisms by which these agents contribute to sarcoidosis development are not fully understood, various reports suggest a causal element related to occupational exposure such as inhalation of World Trade Center dust by fire fighters, or exacerbation of pre-existing disease due to repeated or prolonged exposures. Studies using the Bradford Hill criteria for causation to determine if a causal relationship can be established between occupational exposure and sarcoidosis noted association between World Trade Center (WTC) dust and sarcoidosis, which illustrates a causal relationship, compared to other occupational exposures99. Various occupational settings, such as construction, mining, agriculture, and healthcare, involve exposure to inorganic dusts (e.g., silica, beryllium, aluminum), which have been implicated as potential triggers of sarcoidosis100–102. Using univariable analysis, one group identified elevated risk of sarcoidosis for workers with industrial organic dust exposures, suppliers of building materials, hardware, and gardening materials, as well as educators. Work providing childcare was negatively associated with sarcoidosis risk103. These studies provide valuable insights into occupational exposures that have been associated with sarcoidosis. However, it’s important to note that occupational exposure does not account for all cases of sarcoidosis, and the disease can also occur in individuals without any known occupational risk factors. Further research is needed to better understand the relationship between occupational exposures and sarcoidosis development.
Chronic Beryllium Disease (CBD)-
Similar to most immune-mediated diseases, disease susceptibility in CBD and sarcoidosis is driven by the expression of certain MHCII molecules, primarily HLA-DPB1 in CBD and several HLA-DRB1 alleles in sarcoidosis. One of the defining features of both CBD and sarcoidosis is an infiltration of activated CD4+ T cells in the lung. CD4+ T cells in the bronchoalveolar lavage (BAL) of CBD and sarcoidosis patients are highly Th1 polarized, and there is a significant increase in inflammatory Th1 cytokines present in the BAL fluid. In sarcoidosis, there is also a significant population of Th17 cells in the lungs that is not present in CBD. Due to persistent antigen exposure and chronic inflammation in the lung, these activated CD4+ T cells often display either an exhausted or anergic phenotype. Evidence suggests that these T cells are responding to common antigens in the lung. In CBD there is an expansion of beryllium-responsive TRBV5.1+ TCRs expressed on pathogenic CD4+ T cells derived from the BAL of CBD patients that react with endogenous human peptides derived from the plexin A protein104. performed logistic regression modeling and generated ROC curves to determine which genes could most accurately differentiate: 1) CBD versus sarcoidosis 2) CBD versus BeS 3) sarcoidosis versus controls 4) non-progressive versus progressive sarcoidosis. Results CD55 and TNFα were significantly upregulated, while CXCL9 was significantly downregulated in CBD compared to sarcoidosis (p < 0.05). The ROC curve from the logistic regression model demonstrated high discriminatory ability of the combination of CD55, TNFα, and CXCL9 to distinguish between CBD and sarcoidosis with an AUC of 0.98. CD55 and TNFα were significantly downregulated in sarcoidosis compared to controls (p < 0.05)105.
Inorganic dust.
Inorganic dusts have been implicated in the development of sarcoidosis. While the exact mechanisms are not fully understood, it is believed that the interaction between these inorganic dusts and the immune system drive granuloma formation. Investigators performed MELISA lymphocyte proliferation test on lymphocytes from the blood of 13 sarcoidosis patients using silica and metals, such as aluminum, titanium, nickel, chromium, mercury, and palladium. Of the patient group, nine tested positive to at least one of these metals; two tested positive to beryllium; and two, to silica. Twenty-six metal- or silica-exposed patients with sarcoidosis, seven unexposed patients with sarcoidosis, and 19 control subjects were tested. Only patients with sarcoidosis exhibited immunoreactivity to at least one of the antigens tested (n ¼ 7; P ¼ .039). Correlation between the LPT (ATS protocol) and the MELISA LPT was strong (1.00; P < .01) when testing reactivity to beryllium106. These findings suggest an etiologic mechanism for sarcoidosis similar to that for chronic beryllium disease. Sarcoidosis is also linked to both more subtle metal exposures, such as the copper, iron, and silica in photocopier toner107, as well as a number of vocations that directly handle metals98. Indeed, one study found a significant correlation between exposure to man-made mineral fibers and the development of sarcoidosis. This association was further supported by electron microscopy quantitative analysis on previously collected lung specimens from the sarcoidosis group, which identified silica, aluminum, and/or titanium in 50% of samples108. Despite these associations, one report noted that jobs with metal dust or metal fume exposures were negatively associated with sarcoidosis risk, especially in Caucasian workers103.
Organic Dusts:
Composition of organic dust is very complex, involving particles of microbial, animal and plant origin. Agricultural work, particularly farming and animal husbandry, has been associated with an increased risk of sarcoidosis. Exposure to organic dusts serves as potential triggers of granulomatous inflammation in susceptible individuals35. An investigation assessed the in vitro effects of the co-exposure of fungal cell wall agents (FCWAs) and bacterial lipopolysaccharide (LPS) on inflammatory immune responses of peripheral blood mononuclear cells (PBMCs) from patients with pulmonary sarcoidosis. They reported significantly higher secretion of inflammatory cytokines TNF-α, IL-6, IL-10 and IL-12 (1.7-fold, 2.0-fold, 2.2-fold, and 2.8-fold, respectively; all p < 0.05) after in vitro co-stimulation of PBMCs with FCWAs and LPS35. Because organic dust has a complex composition, future studies to identify the relevant infectious and noninfectious component(s) is warranted.
Tattos-
Cosmetic tattoos despite small size and thereby low relative dose of pigment injected in the skin can trigger fully developed systemic sarcoidosis. It is hypothesized that iron oxide pigments popular in cosmetic tattoo inks of red or brown color may be prone to elicit sarcoid reactions and thus carry a special risk of granuloma. In decorative tattoos, carbon black is the commonest trigger. Both local and systemic sarcoidosis involvement can appear after application of cosmetic tattoos109. Cutaneous, pulmonary and/or ocular involvement in the form of uveitis are among the more common manifestations following cosmetic tattoos. The sarcoidosis lesions induced by tattoos are characterized by increased CD11+ macrophages and CD3+ lymphocytes. 18F-fluorodeoxyglucose (18F-FDG) PET is a nuclear medicine imaging study supports that some sarcoidosis lesions associated with cosmetic tattoos are metabolically over-active areas110. This is one of the first investigations to detail enhanced metabolomics in sarcoidosis granulomas induced by tattoos.
Microbiome
Perturbations to the lung microbial community, such as the introduction of a respiratory virus or induction of variation by disease or antimicrobials, influence pathogenesis. For example, murine models of pulmonary influenza virus infection increase Enterobacteriaceae while reducing Lactobacilli and Lactococci in the intestinal microbial community111.
Lung microbiome:
While a specific microbial agent has not been identified as an etiologic agent of pulmonary sarcoidosis, a growing body of literature supports that a lack of pulmonary microbial diversity is associated with more severe forms of disease. For example, a team of investigators performed a sampling of oral wash (OW), protected bronchoalveolar lavage fluid (PBAL) and left protected sterile brushes (LPSB) of 35 sarcoidosis patients and 35 healthy controls112. The pulmonary microbiome was profiled by sequencing the fungal internal transcribed spacer 1 (ITS1) region and the V3V4 region of the bacterial 16S rRNA gene There was no significant difference in fungal alpha diversity between sarcoidosis and controls, but compared to controls, the bacterial alpha diversity in sarcoidosis was significantly lower in oral wash (p = 0.047) and BAL (p = 0.03). Aspergillus dominated the PBAL samples in sarcoidosis 112.
An independent investigation of BAL from stable COPD patients, exacerbated COPD (ECOPD), interstitial lung disease (ILD) and sarcoidosis was conducted to analyze SSU rRNA gene sequences in order to define the alveolar microbiome. Shannon diversity was significantly higher in stable COPD when compared to ECOPD (p = 0.0061) and ILD (p = 0.037) patient samples. No significant difference in observed richness was noted between the diseases (p = 0.099, Kruskal–Wallis test). A predominance of Proteobacteria followed by Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria was observed in all the disease subsets. However, using Shannon diversity index, statistically significant differences between diseases (p = 0.001, Kruskal–Wallis test) with post hoc tests revealed higher diversity in stable COPD compared to ILD (p = 0.037) and sarcoidosis (p = 0.004); Mann–Whitney test, Bonferroni adjustment). Bray–Curtis based PCA plots were analyzed at the genus level to understand the community ordination. This approach showed extensive overlap in membership between the bacterial communities of the ECOPD, stable COPD, ILD, and sarcoidosis disease groups. These findings suggest that while there is extensive microbial community overlap among chronic pulmonary diseases, quantifiable distinctions in the percentages of microbes vary by disease113. Atopobium and Fusobacterium spp were noted in significantly higher prevalence among sarcoidosis BAL, compared to IPF and healthy control specimens114.
Similarly, another group of investigators performed 16sRNA gene sequencing of BAL obtained from the middle lobe or the lingula of sarcoidosis and ILD patients recruited within the PULMOHOM study, a prospective cohort study to characterize inflammatory processes in pulmonary diseases. The sarcoidosis patients were sorted according to Scadding classification. There were no significant differences between patients with sarcoidosis or other ILDs with regard to microbiome composition and diversity. In addition, the abundance of the genera Atopobium, Fusobacterium, Mycobacterium or Propionibacterium were not different between the two groups 115.
The way the investigations are conducted carry significant implications as bacterial 16S and fungal ITS1 ribosomal RNA gene metagenomic analysis of sarcoidosis specimens, such as formalin-fixed, paraffin-embedded lymph node biopsies, BAL, Kveim reagent, and fresh granulomatous spleen, revealed enrichment of microbes in single types of sarcoidosis samples but limited concordance across sample types. Statistical analysis accounting for environmental contamination was essential to avoiding false positives116. Although the microbiome investigations have not identified an etiologic cause of sarcoidosis, independent investigations support that low microbial diversity contributes to the pathogenesis of fibrotic lung diseases, such as in IPF patients, in which lung bacterial burden predicts fibrosis progression, and microbiota diversity and composition correlate with increased alveolar profibrotic cytokines117.
Gut Microbiome.
The gut microbiome has been shown to play a crucial role in immune system development and homeostasis. Dysbiosis of the gut microbiome, characterized by alterations in the composition and function of the microbiota, has been implicated in the pathogenesis of several autoimmune and inflammatory diseases, including rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis 118.
Another investigation reported the significance of gut microbial diversity to lung fibrosis pathogenesis, reporting the reduced gut microbial diversity leads to increase generation of pulmonary PD-1+Th17 cells, which augments collagen deposition thereby augmenting lung fibrosis severity119. This investigation also demonstrated that the addition of lactobacillus supernatant to human lung fibroblasts reduced the capacity of IL-17A and TGF-β1 to induce collagen 1A levels. These findings suggest that secreted microbial products, rather than the microorganism itself, can directly impact HLF collagen production119. Similar rigorous metagenomic investigations are warranted in sarcoidosis subjects because while independent investigations support a role of microbial dysbiosis in more severe forms of disease, there is a lack of congruence regarding key microbial populations. The dysbiosis of the gut microbiome in sarcoidosis may contribute to the development of granulomas through several mechanisms. First, dysbiosis can lead to alterations in the gut epithelial barrier and increased permeability, allowing for the translocation of bacterial products and antigens into the systemic circulation 120. These antigens can activate the immune system and promote the formation of granulomas. Second, dysbiosis can lead to alterations in the production of short-chain fatty acids, which are important for the maintenance of intestinal homeostasis and immune regulation121. Third, dysbiosis can modulate leukocyte production of cytokines and chemokines, which can affect the recruitment and activation of immune cells122.
Cholesterol
Independent investigations demonstrate significantly increased risks of sarcoidosis among obese compared with nonobese patients; risk estimates ranged from 1.42 [95% confidence interval (CI), 1.07-1.89] to 3.59 (95% CI, 2.31-5.57) 123–125. However, it was unclear if obesity was an inciter, a consequence or both of sarcoidosis emergence; a growing body of literature supports that it contributes to sarcoidosis pathogenesis and emergence. In a cohort of untreated, new diagnosed sarcoidosis subjects, metabolic syndrome was more common in sarcoidosis than controls (odds ratio, OR: 5.3; 95% confidence interval, CI 95%: 2.4-11.5;p<0.001). In addition, triglyceride and glucose levels, diastolic blood pressure measurements, and waist circumference of female sarcoidosis patients were significantly higher than in controls126. Recent studies reveal several mechanisms by which obesity may impact sarcoidosis pathogenesis. Using a sarcoidosis murine model, investigators noted that aberrant lipid metabolism in sarcoidosis macrophages drives lung granuloma formation127. Independent researchers noted increased IL-15 in the sarcoidosis lung of murine model and humans. The presence of obesity was an important driver of IL-15 expression128. In tuberculosis subjects, pathogenic mycobacteria use cholesterol as a carbon source129,130 and cholesterol also serves as a driver of inflammation.
Conclusion
Sarcoidosis is a complex, granulomatous disease with significant clinical and immunologic overlap with known infectious diseases, such as tuberculosis, as well as granulomatous diseases of noninfectious origin, such as CBD. While the etiology(ies) of sarcoidosis remains unknown, it is an exciting time to be a sarcoidosis researcher as significant progress has been achieved in understanding sarcoidosis pathogenesis. The identification of microbial targets of sarcoidosis local and systemic immune response had led to potential therapeutic targets, and the emergence of new sarcoidosis animal models, which further enhance understanding of sarcoidosis pathogenesis. It lends hope that enhanced understanding of how infectious antigens drive sarcoidosis pathogenesis, as well as noninfectious factors such as metals or organic dust, contribute to pathogenesis. Further research is also needed to understand the role of relevant mitigating factors, such as occupational hazards, immunomodulatory medications, the gut microbiome and cholesterol on sarcoidosis progression. Dysbiosis of the gut microbiome may play a role in the pathogenesis of sarcoidosis, with alterations in the composition and function of the microbiota contributing to profibrotic cytokine expression. Further studies are needed to elucidate the mechanisms underlying the gut microbiome-sarcoidosis connection and to determine whether targeting the gut microbiome may be a viable therapeutic strategy for this complex disease.
Figure 1.

Microbial and Non-infectious causes of sarcoidosis pathogenesis
This work provides a detailed list of potential infectious causes.
This works provides a detailed list of non-infectious contributors.
A synopsis figures of potential drives of sarcoidosis progression is also included.
Acknowledgements
This work was supported by grants NIH K24 HL127301-01, and the Ellen Dreiling Research Fund Endowment to W.P.D. O.S.C is supported by T32 Grant T32AR059039-10, and K12 HL 143956-4; Z.W. is supported by T32 HL 087737. We would like to thank Thiagarajan Venkataraman and Aisha Souquette for providing scientific input and editorial assistance. The model was created using BioRender.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interests
The authors declare no competing interests.
References:
- 1.Crouser ED, Maier LA, Wilson KC, et al. Diagnosis and Detection of Sarcoidosis. An Official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2020;201(8):e26–e51. DOI: 10.1164/rccm.202002-0251ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.James DG. The sarcoidosis movement and its personalities. J Med Biogr 1995;3(3):148–60. DOI: 10.1177/096777209500300305. [DOI] [PubMed] [Google Scholar]
- 3.Drake WP, Pei Z, Pride DT, Collins RD, Cover TL, Blaser MJ. Molecular analysis of sarcoidosis tissues for mycobacterium species DNA. Emerg Infect Dis 2002;8(11):1334–41. DOI: 10.3201/eid0811.020318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Oswald-Richter KA, Beachboard DC, Seeley EH, et al. Dual analysis for mycobacteria and propionibacteria in sarcoidosis BAL. J Clin Immunol 2012;32(5):1129–40. DOI: 10.1007/sl0875-012-9700-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Masoud S, Mihan P, Hamed M, Mehdi M, Mohamad RM. The presence of mycobacterial antigens in sarcoidosis associated granulomas. Sarcoidosis Vasc Diffuse Lung Dis 2017;34(3):236–241. DOI: 10.36141/svdld.v34i3.5739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Song Z, Marzilli L, Greenlee BM, et al. Mycobacterial catalase-peroxidase is a tissue antigen and target of the adaptive immune response in systemic sarcoidosis. J Exp Med 2005;201(5):755–67. DOI: 10.1084/jem.20040429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Allen SS, Evans W, Carlisle J, et al. Superoxide dismutase A antigens derived from molecular analysis of sarcoidosis granulomas elicit systemic Th-1 immune responses. Respir Res 2008;9(1):36. DOI: 10.1186/1465-9921-9-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Carlisle J, Evans W, Hajizadeh R, et al. Multiple Mycobacterium antigens induce interferon-gamma production from sarcoidosis peripheral blood mononuclear cells. Clin Exp Immunol 2007;150(3):460–8. DOI: 10.1111/j.l365-2249.2007.03510.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen ES, Wahlstrom J, Song Z, et al. T cell responses to mycobacterial catalase-peroxidase profile a pathogenic antigen in systemic sarcoidosis. J Immunol 2008;181(12):8784–96. DOI: 10.4049/jimmunol.181.12.8784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ostadkarampour M, Eklund A, Moller D, et al. Higher levels of interleukin IL-17 and antigen-specific IL-17 responses in pulmonary sarcoidosis patients with Lofgren’s syndrome. Clin Exp Immunol 2014;178(2):342–52. DOI: 10.1111/cei.l2403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dubaniewicz A. Mycobacterial Heat Shock Proteins in Sarcoidosis and Tuberculosis. Int J Mol Sci 2023;24(6). DOI: 10.3390/ijms24065084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Swaisgood CM, Oswald-Richter K, Moeller SD, et al. Development of a sarcoidosis murine lung granuloma model using Mycobacterium superoxide dismutase A peptide. Am J Respir Cell Mol Biol 2011;44(2):166–74. DOI: 10.1165/rcmb.2009-03500C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Huang Y, Lu CZ, Wang CC, Jiang Y, Wang XY, Duan YY. [Establishment and identification of a C57B/6 mouse sarcoidosis granuloma model]. Zhonghua Jie He He Hu Xi Za Zhi 2013;36(8):587–91. (https://www.ncbi.nlm.nih.gov/pubmed/24252735). [PubMed] [Google Scholar]
- 14.Zhang X, Dai Q, Shan J, et al. Inhibition of phosphoinositide-3 kinases gamma/delta ameliorates pulmonary granuloma by rescuing Treg function in a sarcoidosis model. Exp TherMed 2023;25(5):225. DOI: 10.3892/etm.2023.11923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Drake WP, Oswald-Richter K, Richmond BW, et al. Oral antimycobacterial therapy in chronic cutaneous sarcoidosis: a randomized, single-masked, placebo-controlled study. JAMA Dermatol 2013;149(9):1040–9. DOI: 10.1001/jamadermatol.2013.4646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Drake WP, Culver DA, Baughman RP, et al. Phase II Investigation of the Efficacy of Antimycobacterial Therapy in Chronic Pulmonary Sarcoidosis. Chest 2021;159(5):1902–1912. DOI: 10.1016/j.chest.2020.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Apostu M, Mihaescu T. [Respiratory functional changes in pulmonary tuberculosis]. Pneumologia 2013;62(3):148–57. (https://www.ncbi.nlm.nih.gov/pubmed/24273998). [PubMed] [Google Scholar]
- 18.Park HY, Jeong BH, Chon HR, Jeon K, Daley CL, Koh WJ. Lung Function Decline According to Clinical Course in Nontuberculous Mycobacterial Lung Disease. Chest 2016;150(6):1222–1232. DOI: 10.1016/j.chest.2016.06.005. [DOI] [PubMed] [Google Scholar]
- 19.Yu X, Xie J. Roles and underlying mechanisms of ESAT-6 in the context of Mycobacterium tuberculosis-host interaction from a systems biology perspective. Cell Signal 2012;24(9):1841–6. DOI: 10.1016/j.cellsig.2012.05.014. [DOI] [PubMed] [Google Scholar]
- 20.Liu C, Zhao Z, Fan J, et al. Quantification of circulating Mycobacterium tuberculosis antigen peptides allows rapid diagnosis of active disease and treatment monitoring. Proc Natl Acad Sci U S A 2017;114(15):3969–3974. DOI: 10.1073/pnas.l621360114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mattos AM, Almeida Cde S, Franken KL, et al. Increased IgG1, IFN-gamma, TNF-alpha and IL-6 responses to Mycobacterium tuberculosis antigens in patients with tuberculosis are lower after chemotherapy. Int Immunol 2010;22(9):775–82. DOI: 10.1093/intimm/dxq429. [DOI] [PubMed] [Google Scholar]
- 22.Clifford V, Tebruegge M, Zufferey C, et al. Mycobacteria-specific cytokine responses as correlates of treatment response in active and latent tuberculosis. J Infect 2017;75(2):132–145. DOI: 10.1016/j.jinf.2017.04.011. [DOI] [PubMed] [Google Scholar]
- 23.Yamaguchi T, Costabel U, McDowell A, et al. Immunohistochemical Detection of Potential Microbial Antigens in Granulomas in the Diagnosis of Sarcoidosis. J Clin Med 2021;10(5). DOI: 10.3390/jcml0050983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Uchida K, Furukawa A, Yoneyama A, et al. Propionibacterium acnes-Derived Circulating Immune Complexes in Sarcoidosis Patients. Microorganisms 2021;9(11). DOI: 10.3390/microorganisms9112194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Akata K, Yamasaki K, Nemoto K, et al. Sarcoidosis Associated with Enlarged Mediastinal Lymph Nodes with the Detection of Streptococcus gordonii and Cutibacterium acnes Using a Clone Library Method. Intern Med 2024;63(2):299–304. DOI: 10.2169/internaImedicine.1887-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jiang D, Xiao H, Zheng X, Dong R, Zhang H, Dai H. Interleukin-17A plays a key role in pulmonary fibrosis following Propionibacterium acnes-induced sarcoidosis-like inflammation. Exp Biol Med (Maywood) 2023;248(14):1181–1190. DOI: 10.1177/15353702231182224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Biberfeld P, Petren AL, Eklund A, et al. Human herpesvirus-6 (HHV-6, HBLV) in sarcoidosis and lymphoproliferative disorders. J Virol Methods 1988;21(1-4):49–59. DOI: 10.1016/0166-0934(88)90051-1. [DOI] [PubMed] [Google Scholar]
- 28.Rottoli P, Bianchi Bandinelli ML, Rottoli L, Zazzi M, Panzardi G, Valensin PE. Sarcoidosis and infections by human lymphotropic viruses. Sarcoidosis 1990;7(1):31–3. (https://www.ncbi.nlm.nih.gov/pubmed/2161122). [PubMed] [Google Scholar]
- 29.Brito-Zeron P, Gracia-Tello B, Robles A, et al. Characterization and Outcomes of SARS-CoV-2 Infection in Patients with Sarcoidosis. Viruses 2021;13(6). DOI: 10.3390/vl3061000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Vagts C, Sweis JJG, Sweis NWG, et al. Initial behaviors and attitudes towards the COVID-19 vaccine in sarcoidosis patients: results of a self-reporting questionnaire. Sarcoidosis Vasc Diffuse Lung Dis 2023;40(2):e2023012. DOI: 10.36141/svdld.v40i2.14388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Boyton RJ, Altmann DM, Wright A, Kon OM. Pulmonary infection with Cryptococcus neoformans in the face of underlying sarcoidosis. Respiration 2007;74(4):462–6. DOI: 10.1159/000087461. [DOI] [PubMed] [Google Scholar]
- 32.Alloway JA, Buchsbaum RM, Filipov PT, Reynolds BN, Day JA. Mucormycosis in a patient with sarcoidosis. Sarcoidosis 1995;12(2):143–6. (https://www.ncbi.nlm.nih.gov/pubmed/8532963). [PubMed] [Google Scholar]
- 33.Lord GP. Pulmonary sarcoidosis complicated by cryptococcosis and coccidioidomycosis. The changing spectrum of fungus disease in Maine. J Maine Med Assoc 1974;65(10):236–40. (https://www.ncbi.nlm.nih.gov/pubmed/4430873). [PubMed] [Google Scholar]
- 34.Greaves SA, Ravindran A, Santos RG, et al. CD4+ T cells in the lungs of acute sarcoidosis patients recognize an Aspergillus nidulans epitope. J Exp Med 2021;218(10). DOI: 10.1084/jem.20210785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Stopinsek S, Ihan A, Salobir B, Tercelj M, Simcic S. Fungal cell wall agents and bacterial lipopolysaccharide in organic dust as possible risk factors for pulmonary sarcoidosis. J Occup Med Toxicol 2016;11:46. DOI: 10.1186/sl2995-016-0135-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mortaz E, Adcock IM, Abedini A, et al. The role of pattern recognition receptors in lung sarcoidosis. Eur J Pharmacol 2017;808:44–48. DOI: 10.1016/j.ejphar.2017.01.020. [DOI] [PubMed] [Google Scholar]
- 37.Krausgruber T, Redl A, Barreca D, et al. Single-cell and spatial transcriptomics reveal aberrant lymphoid developmental programs driving granuloma formation. Immunity 2023;56(2):289–306 e7. DOI: 10.1016/j.immuni.2023.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hawkins C, Shaginurova G, Shelton DA, et al. Local and Systemic CD4(+) T Cell Exhaustion Reverses with Clinical Resolution of Pulmonary Sarcoidosis. J Immunol Res 2017;2017:3642832. DOI: 10.1155/2017/3642832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Celada LJ, Rotsinger JE, Young A, et al. Programmed Death-1 Inhibition of Phosphatidylinositol 3-Kinase/AKT/Mechanistic Target of Rapamycin Signaling Impairs Sarcoidosis CD4(+) T Cell Proliferation. Am J Respir Cell Mol Biol 2017;56(1):74–82. DOI: 10.1165/rcmb.2016-00370C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Locke LW, Schlesinger LS, Crouser ED. Current Sarcoidosis Models and the Importance of Focusing on the Granuloma. Front Immunol 2020;11:1719. DOI: 10.3389/fimmu.2020.01719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Belyaeva IV, Kosova AN, Vasiliev AG. Tuberculosis and Autoimmunity. Pathophysiology 2022;29(2):298–318. DOI: 10.3390/pathophysiology29020022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dlugovitzky D, Luchesi S, Torres-Morales A, et al. Circulating immune complexes in patients with advanced tuberculosis and their association with autoantibodies and reduced CD4+ lymphocytes. Braz J Med Biol Res 1995;28(3):331–5. (https://www.ncbi.nlm.nih.gov/pubmed/8520527). [PubMed] [Google Scholar]
- 43.Pradhan V, Patwardhan M, Athavale A, Taushid S, Ghosh K. Mycobacterium tuberculosis triggers autoimmunity? Indian J Tuberc 2012;59(1):49–51. (https://www.ncbi.nlm.nih.gov/pubmed/22670513). [PubMed] [Google Scholar]
- 44.Shen CY, Hsieh SC, Yu CL, Wang JY, Lee LN, Yu CJ. Autoantibody prevalence in active tuberculosis: reactive or pathognomonic? BMJ Open 2013;3(7). DOI: 10.1136/bmjopen-2013-002665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Starshinova A, Malkova A, Zinchenko Y, et al. Identification of autoimmune markers in pulmonary tuberculosis. Front Immunol 2022;13:1059714. DOI: 10.3389/fimmu.2022.1059714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shi TY, Wen XH, Shi XH, Meng J, Lu YW. Associations between sarcoidosis, autoimmune diseases, and autoantibodies: a single-center retrospective study in China. Clin Exp Med 2022;22(2):277–283. DOI: 10.1007/sl0238-021-00737-5. [DOI] [PubMed] [Google Scholar]
- 47.Tana C, Drent M, Nunes H, et al. Comorbidities of sarcoidosis. Ann Med 2022;54(1):1014–1035. DOI: 10.1080/07853890.2022.2063375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Murthi M, Yoshioka K, Cho JH, et al. Presence of concurrent sarcoid-like granulomas indicates better survival in cancer patients: a retrospective cohort study. ERJ Open Res 2020;6(4). DOI: 10.1183/23120541.00061-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bagavant H, Cizio K, Araszkiewicz AM, et al. Systemic immune response to vimentin and granuloma formation in a model of pulmonary sarcoidosis. J Transl Autoimmun 2022;5:100153. DOI: 10.1016/j.jtauto.2022.100153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kinloch AJ, Kaiser Y, Wolfgeher D, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Musaelyan A, Lapin S, Nazarov V, et al. Vimentin as antigenic target in autoimmunity: A comprehensive review. Autoimmun Rev 2018;17(9):926–934. DOI: 10.1016/j.autrev.2018.04.004. [DOI] [PubMed] [Google Scholar]
- 52.Hrycaj P, Wurm K, Mennet P, Muller W. Antibodies to heat shock proteins in patients with pulmonary sarcoidosis. Sarcoidosis 1995;12(2):124–30. (https://www.ncbi.nlm.nih.gov/pubmed/8532959). [PubMed] [Google Scholar]
- 53.Spagnolo P, Sato H, Marshall SE, et al. Association between heat shock protein 70/Hom genetic polymorphisms and uveitis in patients with sarcoidosis. Invest Ophthalmol Vis Sci 2007;48(7):3019–25. DOI: 10.1167/iovs.06-1485. [DOI] [PubMed] [Google Scholar]
- 54.Chen ES. Innate immunity in sarcoidosis pathobiology. Curr Opin Pulm Med 2016;22(5):469–75. DOI: 10.1097/MCP.0000000000000305. [DOI] [PubMed] [Google Scholar]
- 55.Chen ES, Song Z, Willett MH, et al. Serum amyloid A regulates granulomatous inflammation in sarcoidosis through Toll-like receptor-2. Am J Respir Crit Care Med 2010;181(4):360–73. DOI: 10.1164/rccm.200905-06960C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Beijer E, Roodenburg-Benschop C, Schimmelpennink MC, Grutters JC, Meek B, Veltkamp M. Elevated Serum Amyloid a Levels Are not Specific for Sarcoidosis but Associate with a Fibrotic Pulmonary Phenotype. Cells 2021;10(3). DOI: 10.3390/cellsl0030585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rubio-Rivas M, Moreira C, Marcoval J. Sarcoidosis related to checkpoint and BRAF/MEK inhibitors in melanoma. Autoimmun Rev 2020;19(8):102587. DOI: 10.1016/j.autrev.2020.102587. [DOI] [PubMed] [Google Scholar]
- 58.Chopra A, Nautiyal A, Kalkanis A, Judson MA. Drug-Induced Sarcoidosis-Like Reactions. Chest 2018;154(3):664–677. DOI: 10.1016/j.chest.2018.03.056. [DOI] [PubMed] [Google Scholar]
- 59.Handa T, Nagai S, Ito I, et al. Cytotoxic T-lymphocyte antigen-4 (CTLA-4) exon 1 polymorphism affects lymphocyte profiles in bronchoalveolar lavage of patients with sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2003;20(3):190–6. (https://www.ncbi.nlm.nih.gov/pubmed/14620161). [PubMed] [Google Scholar]
- 60.Celada LJ, Kropski JA, Herazo-Maya JD, et al. PD-1 up-regulation on CD4(+) T cells promotes pulmonary fibrosis through STAT3-mediated IL-17A and TGF-beta1 production. Sci Transl Med 2018;10(460). DOI: 10.1126/scitranslmed.aar8356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Braun NA, Celada LJ, Herazo-Maya JD, et al. Blockade of the programmed death-1 pathway restores sarcoidosis CD4(+) T-cell proliferative capacity. Am J Respir Crit Care Med 2014;190(5):560–71. DOI: 10.1164/rccm.201401-01880C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kim ST, Pundole X, Dadu R, Lambotte O, Ramos-Casals M, Suarez-Almazor ME. Use of immune checkpoint inhibitors in cancer patients with pre-existing sarcoidosis. Immunotherapy 2021;13(6):465–475. DOI: 10.2217/imt-2020-0272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Broos CE, Koth LL, van Nimwegen M, et al. Increased T-helper 17.1 cells in sarcoidosis mediastinal lymph nodes. Eur Respir J 2018;51(3). DOI: 10.1183/13993003.01124-2017. [DOI] [PubMed] [Google Scholar]
- 64.Lomax AJ, McGuire HM, McNeil C, et al. Immunotherapy-induced sarcoidosis in patients with melanoma treated with PD-1 checkpoint inhibitors: Case series and immunophenotypic analysis. Int J Rheum Dis 2017;20(9):1277–1285. DOI: 10.1111/1756-185X.13076. [DOI] [PubMed] [Google Scholar]
- 65.Li Y, Flavell RR, Juarez R, et al. Retrospective study of the incidence of sarcoidosis-like reaction in patients treated with immunotherapy. Clin Radiol 2023;78(2):e131–e136. DOI: 10.1016/j.crad.2022.09.127. [DOI] [PubMed] [Google Scholar]
- 66.Li H, Mu F, Zou B, Wang L. Pulmonary sarcoidosis-like reactions induced by sintilimab in esophageal cancer: A case report. Medicine (Baltimore) 2023;102(31):e34432. DOI: 10.1097/MD.0000000000034432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Morris DG, Jasmer RM, Huang L, Gotway MB, Nishimura S, King TE Jr. Sarcoidosis following HIV infection: evidence for CD4+ lymphocyte dependence. Chest 2003;124(3):929–35. DOI: 10.1378/chest.124.3.929. [DOI] [PubMed] [Google Scholar]
- 68.Miranda EJ, Leite OH, Duarte MI. Immune reconstitution inflammatory syndrome associated with pulmonary sarcoidosis in an HIV-infected patient: an immunohistochemical study. Braz J Infect Dis 2011;15(6):601–6. DOI: 10.1590/sl413-86702011000600018. [DOI] [PubMed] [Google Scholar]
- 69.Foulon G, Wislez M, Naccache JM, et al. Sarcoidosis in HIV-infected patients in the era of highly active antiretroviral therapy. Clin Infect Dis 2004;38(3):418–25. DOI: 10.1086/381094. [DOI] [PubMed] [Google Scholar]
- 70.Marzouk K, Saleh S, Kannass M, Sharma OP. Interferon-induced granulomatous lung disease. Curr Opin Pulm Med 2004;10(5):435–40. DOI: 10.1097/01.mcp.0000134400.88832.9c. [DOI] [PubMed] [Google Scholar]
- 71.CI Rodrigues-Fernandes, Migliorati CA, Gueiros LAM, et al. Adalimumab-induced sarcoidosis-like reaction involving oral cavity in rheumatoid arthritis: a case-based review. Clin Rheumatol 2021;40(9):3833–3839. DOI: 10.1007/sl0067-021-05654-2. [DOI] [PubMed] [Google Scholar]
- 72.Salvatierra J, Magro-Checa C, Rosales-Alexander JL, Raya-Alvarez E. Acute sarcoidosis as parotid fever in rheumatoid arthritis under anti-tumor necrosis factor-alpha therapy. Rheumatology (Oxford) 2011;50(7):1346–8. DOI: 10.1093/rheumatology/kerl66. [DOI] [PubMed] [Google Scholar]
- 73.Sharp M, Donnelly SC, Moller DR. Tocilizumab in sarcoidosis patients failing steroid sparing therapies and anti-TNF agents. Respir Med X 2019;1. DOI: 10.1016/j.yrmex.2019.100004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lambert N, Hansen I, El Moussaoui M, et al. Lung and liver sarcoidosis-like reaction induced by tocilizumab. Br J Clin Pharmacol 2021;87(12):4848–4852. DOI: 10.1111/bcp.14878. [DOI] [PubMed] [Google Scholar]
- 75.Baughman RP, Teirstein AS, Judson MA, et al. Clinical characteristics of patients in a case control study of sarcoidosis. Am J Respir Crit Care Med 2001;164(10 Pt 1):1885–9. DOI: 10.1164/ajrccm.164.10.2104046. [DOI] [PubMed] [Google Scholar]
- 76.Baker MC, Vago E, Tamang S, Horvath-Puho E, Sorensen HT. Sarcoidosis rates in BCG-vaccinated and unvaccinated young adults: A natural experiment using Danish registers. Semin Arthritis Rheum 2023;60:152205. DOI: 10.1016/j.semarthrit.2023.152205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sutherland I, Mitchell DN, Hart PD. Incidence of Intrathoracic Sarcoidosis among Young Adults Participating in a Trial of Tuberculosis Vaccines. Br Med J 1965;2(5460):497–503. DOI: 10.1136/bmj.2.5460.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sutherland I. Mycobacteria and sarcoidosis. BCG ineffective against sarcoidosis. BMJ 1993;306(6887):1270; author reply 1270-1. DOI: 10.1136/bmj.306.6887.1270-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kawabata A, Nakamura T, Suzuki H, Yoshida M, Otsuki Y, Mori N. Coincidence of Sarcoidosis and a COVID-19 vaccine-associated hypermetabolic lymphadenopathy in a patient with a history of invasive breast cancer: A case report. Int J Surg Case Rep 2022;94:107098. DOI: 10.1016/j.ijscr.2022.107098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Niebel D, Novak N, Wilhelmi J, et al. Cutaneous Adverse Reactions to COVID-19 Vaccines: Insights from an Immuno-Dermatological Perspective. Vaccines (Basel) 2021;9(9). DOI: 10.3390/vaccines9090944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Abu Rached N, Mansour R, Susok L, et al. Sarcoidal immune reaction following SARS-CoV-2 vaccination. Clin Exp Dermatol 2022;47(5):970–972. DOI: 10.1111/ced.15082. [DOI] [PubMed] [Google Scholar]
- 82.Rodriguez Y, Rojas M, Beltran S, et al. Autoimmune and autoinflammatory conditions after COVID-19 vaccination. New case reports and updated literature review. J Autoimmun 2022;132:102898. DOI: 10.1016/j.jaut.2022.102898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Barzo P, Tamasi L. [Lofgren syndrome after silicone breast prosthesis implantation]. Orv Hetil 1998;139(39):2323–6. (https://www.ncbi.nlm.nih.gov/pubmed/9789987). [PubMed] [Google Scholar]
- 84.Alijotas-Reig J, Hindie M, Kandhaya-Pillai R, Miro-Mur F. Bioengineered hyaluronic acid elicited a nonantigenic T cell activation: implications from cosmetic medicine and surgery to nanomedicine. J Biomed Mater Res A 2010;95(1):180–90. DOI: 10.1002/jbm.a.32794. [DOI] [PubMed] [Google Scholar]
- 85.Vera-Lastra O, Medina G, Cruz-Dominguez Mdel P, et al. Human adjuvant disease induced by foreign substances: a new model of ASIA (Shoenfeld’s syndrome). Lupus 2012;21(2):128–35. DOI: 10.1177/0961203311429317. [DOI] [PubMed] [Google Scholar]
- 86.Chang KC, Chan KT, Chong LY, Lau KS, Tam CM, Lam CW. Cutaneous and pulmonary sarcoidosis in a Hong Kong Chinese woman with silicone breast prostheses. Respirology 2003;8(3):379–82. DOI: 10.1046/j.1440-1843.2003.00447.x. [DOI] [PubMed] [Google Scholar]
- 87.Yoshida T, Tanaka M, Okamoto K, Hirai S. Neurosarcoidosis following augmentation mammoplasty with silicone. Neurol Res 1996;18(4):319–20. DOI: 10.1080/01616412.1996.11740428. [DOI] [PubMed] [Google Scholar]
- 88.Redondo P, Del Olmo J, Alberola I. In situ and distant foreign body granulomas caused by silicone. Treatment with allopurinol. Br J Dermatol 2005;152(5):1064–5. DOI: 10.1111/j.1365-2133.2005.06525.x. [DOI] [PubMed] [Google Scholar]
- 89.Teuber SS, Howell LP, Yoshida SH, Gershwin ME. Remission of sarcoidosis following removal of silicone gel breast implants. Int Arch Allergy Immunol 1994;105(4):404–7. DOI: 10.1159/000236791. [DOI] [PubMed] [Google Scholar]
- 90.Balbouzis T, Georgiadis T, Grigoris P. Granulomatous Lung Disease: A Novel Complication following Metallosis from Hip Arthroplasty. Hip Pelvis 2016;28(4):249–253. DOI: 10.5371/hp.2016.28.4.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Jacobs JJ, Urban RM, Wall J, Black J, Reid JD, Veneman L. Unusual foreign-body reaction to a failed total knee replacement: simulation of a sarcoma clinically and a sarcoid histologically. A case report. J Bone Joint Surg Am 1995;77(3):444–51. DOI: 10.2106/00004623-199503000-00015. [DOI] [PubMed] [Google Scholar]
- 92.Prezant DJ, Dhala A, Goldstein A, et al. The incidence, prevalence, and severity of sarcoidosis in New York City firefighters. Chest 1999;116(5):1183–93. DOI: 10.1378/chest.116.5.1183. [DOI] [PubMed] [Google Scholar]
- 93.Reich JM. Sarcoidosis in World Trade Center-Exposed Firefighters. Chest 2018;153(4):1072–1073. DOI: 10.1016/j.chest.2017.11.045. [DOI] [PubMed] [Google Scholar]
- 94.Edmondstone WM. Sarcoidosis in nurses: is there an association? Thorax 1988;43(4):342–3. DOI: 10.1136/thx.43.4.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Pietinalho A, Ohmichi M, Hirasawa M, Hiraga Y, Lofroos AB, Selroos O. Familial sarcoidosis in Finland and Hokkaido, Japan--a comparative study. Respir Med 1999;93(6):408–12. DOI: 10.1053/rmed.1999.0579. [DOI] [PubMed] [Google Scholar]
- 96.Kirschner RA Jr., Parker BC, Falkinham JO 3rd. Epidemiology of infection by nontuberculous mycobacteria. Mycobacterium avium, Mycobacterium intracellulare, and States and their association with environmental variables. Am Rev Respir Dis 1992;145(2 Pt 1):271–5. DOI: 10.1164/ajrccm/145.2_Pt_1.271. [DOI] [PubMed] [Google Scholar]
- 97.Iivanainen E, Martikainen PJ, Vaananen P, Katila ML. Environmental factors affecting the occurrence of mycobacteria in brook sediments. J Appl Microbiol 1999;86(4):673–81. DOI: 10.1046/j.1365-2672.1999.00711.x. [DOI] [PubMed] [Google Scholar]
- 98.Judson MA. Environmental Risk Factors for Sarcoidosis. Frontiers in Immunology 2020;11:1340. DOI: 10.3389/fimmu.2020.01340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lin NW, Maier LA. Occupational exposures and sarcoidosis: current understanding and knowledge gaps. Curr Opin Pulm Med 2022;28(2):144–151. DOI: 10.1097/MCP.0000000000000835. [DOI] [PubMed] [Google Scholar]
- 100.Newman KL, Newman LS. Occupational causes of sarcoidosis. Curr Opin Allergy Clin Immunol 2012;12(2):145–50. DOI: 10.1097/ACI.0b013e3283515173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Izbicki G, Chavko R, Banauch GI, et al. World Trade Center “sarcoid-like” granulomatous pulmonary disease in New York City Fire Department rescue workers. Chest 2007;131(5):1414–23. DOI: 10.1378/chest.06-2114. [DOI] [PubMed] [Google Scholar]
- 102.Hnizdo E, Vallyathan V. Chronic obstructive pulmonary disease due to occupational exposure to silica dust: a review of epidemiological and pathological evidence. Occup Environ Med 2003;60(4):237–43. DOI: 10.1136/oem.60.4.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Barnard J, Rose C, Newman L, et al. Job and industry classifications associated with sarcoidosis in A Case-Control Etiologic Study of Sarcoidosis (ACCESS). J Occup Environ Med 2005;47(3):226–34. DOI: 10.1097/01.jom.0000155711.88781.91. [DOI] [PubMed] [Google Scholar]
- 104.Greaves SA, Atif SM, Fontenot AP. Adaptive Immunity in Pulmonary Sarcoidosis and Chronic Beryllium Disease. Front Immunol 2020;11:474. DOI: 10.3389/fimmu.2020.00474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lin NW, Maier LA, Mroz MM, et al. Genomic biomarkers in chronic beryllium disease and sarcoidosis. Respir Med 2021;187:106390. DOI: 10.1016/j.rmed.2021.106390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Oliver LC, Zarnke AM. Sarcoidosis: An Occupational Disease? Chest 2021;160(4):1360–1367. DOI: 10.1016/j.chest.2021.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Waphare BN, Joshi V. Characterizations of Standard Elements in Posets. Order 2004;21(1):49–60. DOI: 10.1007/sll083-004-2862-x. [DOI] [Google Scholar]
- 108.Drent M, Bomans PH, Van Suylen RJ, Lamers RJ, Bast A, Wouters EF. Association of manmade mineral fibre exposure and sarcoidlike granulomas. Respir Med 2000;94(8):815–20. DOI: 10.1053/rmed.2000.0827. [DOI] [PubMed] [Google Scholar]
- 109.Kanakamedala AD, Maamari RN, Couch SM. Tattoo-associated lacrimal gland enlargement and sarcoidosis. Am J Ophthalmol Case Rep 2023;32:101889. DOI: 10.1016/j.ajoc.2023.101889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Klarskov CK, Danebod K, Brinth LS, Dreyer M. [Not Available]. Ugeskr Laeger 2023;185(33) (https://www.ncbi.nlm.nih.gov/pubmed/37615154). [PubMed] [Google Scholar]
- 111.Groves HT, Cuthbertson L, James P, Moffatt MF, Cox MJ, Tregoning JS. Respiratory Disease following Viral Lung Infection Alters the Murine Gut Microbiota. Front Immunol 2018;9:182. DOI: 10.3389/fimmu.2018.00182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Knudsen KS, Lehmann S, Nielsen R, et al. The lower airways microbiota and antimicrobial peptides indicate dysbiosis in sarcoidosis. Microbiome 2022;10(1):175. DOI: 10.1186/s40168-022-01362-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gupta S, Shariff M, Chaturvedi G, et al. Comparative analysis of the alveolar microbiome in COPD, ECOPD, Sarcoidosis, and ILD patients to identify respiratory illnesses specific microbial signatures. Sci Rep 2021;11(1):3963. DOI: 10.1038/s41598-021-83524-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zimmermann A, Knecht H, Hasler R, et al. Atopobium and Fusobacterium as novel candidates for sarcoidosis-associated microbiota. Eur Respir J 2017;50(6). DOI: 10.1183/13993003.00746-2016. [DOI] [PubMed] [Google Scholar]
- 115.Becker A, Vella G, Galata V, et al. The composition of the pulmonary microbiota in sarcoidosis - an observational study. Respir Res 2019;20(1):46. DOI: 10.1186/sl2931-019-1013-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Clarke EL, Lauder AP, Hofstaedter CE, et al. Microbial Lineages in Sarcoidosis. A Metagenomic Analysis Tailored for Low-Microbial Content Samples. Am J Respir Crit Care Med 2018;197(2):225–234. DOI: 10.1164/rccm.201705-08910C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.O’Dwyer DN, Ashley SL, Gurczynski SJ, et al. Lung Microbiota Contribute to Pulmonary Inflammation and Disease Progression in Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;199(9):1127–1138. DOI: 10.1164/rccm.201809-16500C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Mousa WK, Chehadeh F, Husband S. Microbial dysbiosis in the gut drives systemic autoimmune diseases. Front Immunol 2022;13:906258. DOI: 10.3389/fimmu.2022.906258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chioma OS, Mallott EK, Chapman A, et al. Gut microbiota modulates lung fibrosis severity following acute lung injury in mice. Commun Biol 2022;5(1):1401. DOI: 10.1038/s42003-022-04357-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Fasano A. Intestinal permeability and its regulation by zonulin: diagnostic and therapeutic implications. Clin Gastroenterol Hepatol 2012;10(10):1096–100. DOI: 10.1016/j.cgh.2012.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16(6):341–52. DOI: 10.1038/nri.2016.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157(1):121–41. DOI: 10.1016/j.cell.2014.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Cozier YC, Govender P, Berman JS. Obesity and sarcoidosis: consequence or contributor? Curr Opin Pulm Med 2018;24(5):487–494. DOI: 10.1097/MCP.0000000000000503. [DOI] [PubMed] [Google Scholar]
- 124.Ungprasert P, Crowson CS, Matteson EL. Smoking, obesity and risk of sarcoidosis: A population-based nested case-control study. Respir Med 2016;120:87–90. DOI: 10.1016/j.rmed.2016.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Dumas O, Boggs KM, Cozier YC, Stampfer MJ, Camargo CA Jr., Prospective study of body mass index and risk of sarcoidosis in US women. Eur Respir J 2017;50(4). DOI: 10.1183/13993003.01397-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Isik AC, Kavas M, Tezcan ME. Metabolic syndrome may be more frequent in treatment-naive sarcoidosis patients. Z Rheumatol 2022. DOI: 10.1007/s00393-022-01210-8. [DOI] [PubMed] [Google Scholar]
- 127.Lim CX, Redl A, Kleissl L, et al. Aberrant Lipid Metabolism in Macrophages Is Associated with Granuloma Formation in Sarcoidosis. Am J Respir Crit Care Med 2024. DOI: 10.1164/rccm.202307-12730C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Minasyan M, Sharma L, Pivarnik T, et al. Elevated IL-15 concentrations in the sarcoidosis lung are independent of granuloma burden and disease phenotypes. Am J Physiol Lung Cell Mol Physiol 2021;320(6):L1137–L1146. DOI: 10.1152/ajplung.00575.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nazarova EV, Montague CR, La T, et al. Rv3723/LucA coordinates fatty acid and cholesterol uptake in Mycobacterium tuberculosis. Elife 2017;6. DOI: 10.7554/eLife.26969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Babunovic GH, DeJesus MA, Bosch B, et al. CRISPR Interference Reveals That All-Trans-Retinoic Acid Promotes Macrophage Control of Mycobacterium tuberculosis by Limiting Bacterial Access to Cholesterol and Propionyl Coenzyme A. mBio 2022;13(1):e0368321. DOI: 10.1128/mbio.03683-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Garzoni C, Brugger SD, Qi W, et al. Microbial communities in the respiratory tract of patients with interstitial lung disease. Thorax 2013;68(12):1150–6. DOI: 10.1136/thoraxjnl-2012-202917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yousuf H, Mekki R, Khan K, Hussain A. Pembrolizumab-Induced Sarcoid-Like Reaction in a Patient With Lung Cancer. Cureus 2020;12(12):e12395. DOI: 10.7759/cureus.12395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Nishino M, Sholl LM, Hodi FS, Hatabu H, Ramaiya NH. Anti-PD-1-Related Pneumonitis during Cancer Immunotherapy. N Engl J Med 2015;373(3):288–90. DOI: 10.1056/NEJMcl505197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Cheshire SC, Board RE, Lewis AR, Gudur LD, Dobson MJ. Pembrolizumab-induced Sarcoidlike Reactions during Treatment of Metastatic Melanoma. Radiology 2018;289(2):564–567. DOI: 10.1148/radiol.2018180572. [DOI] [PubMed] [Google Scholar]
- 135.Reddy SB, Possick JD, Kluger HM, Galan A, Han D. Sarcoidosis Following Anti-PD-1 and Anti-CTLA-4 Therapy for Metastatic Melanoma. J Immunother 2017;40(8):307–311. DOI: 10.1097/CJ1.0000000000000181. [DOI] [PubMed] [Google Scholar]
- 136.Cho HS, Kim SJ, Yoo JY. Sarcoidosis during treatment of pulmonary tuberculosis: a rare case report and review of the literature. J Int Med Res 2021;49(4):3000605211001632. DOI: 10.1177/03000605211001632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.van Enschot JW, van Balkom RH. Sarcoidosis following Mycobacterium tuberculosis infection: Coincidence or consequence. Respir Med Case Rep 2013;9:11–4. DOI: 10.1016/j.rmcr.2013.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ravenel JG, McAdams HP, Plankeel JF, Butnor KJ, Sporn TA. Sarcoidosis induced by interferon therapy. AJR Am J Roentgenol 2001;177(1):199–201. DOI: 10.2214/ajr.177.1.1770199. [DOI] [PubMed] [Google Scholar]
- 139.Gianella F, Hsia CC, Sakhaee K. The role of vitamin D in sarcoidosis. Fac Rev 2020;9:14. DOI: 10.12703/b/9-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Kiani A, Abedini A, Adcock IM, et al. Association Between Vitamin D Deficiencies in Sarcoidosis with Disease Activity, Course of Disease and Stages of Lung Involvements. J Med Biochem 2018;37(2):103–109. DOI: 10.1515/jomb-2017-0041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ungprasert P, Crowson CS, Matteson EL. Seasonal variation in incidence of sarcoidosis: a population-based study, 1976-2013. Thorax 2016;71(12):1164–1166. DOI: 10.1136/thoraxjnl-2016-209032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hosoda Y, Sasagawa S, Yamaguchi T. Sarcoidosis and tuberculosis: epidemiological similarities and dissimilarities. A review of a series of studies in a Japanese work population (1941-1996) and the general population (1959-1984). Sarcoidosis Vasc Diffuse Lung Dis 2004;21(2):85–93. (https://www.ncbi.nlm.nih.gov/pubmed/15281429). [PubMed] [Google Scholar]
- 143.Judson MA. Environmental Risk Factors for Sarcoidosis. Front Immunol 2020;11:1340. DOI: 10.3389/fimmu.2020.01340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Arkema EV, Grunewald J, Kullberg S, Eklund A, Askling J. Sarcoidosis incidence and prevalence: a nationwide register-based assessment in Sweden. Eur Respir J 2016;48(6):1690–1699. DOI: 10.1183/13993003.00477-2016. [DOI] [PubMed] [Google Scholar]
- 145.Baughman RP, Field S, Costabel U, et al. Sarcoidosis in America. Analysis Based on Health Care Use. Ann Am Thorac Soc 2016;13(8):1244–52. DOI: 10.1513/AnnalsATS.201511-760OC. [DOI] [PubMed] [Google Scholar]
- 146.Dumas O, Abramovitz L, Wiley AS, Cozier YC, Camargo CA Jr. Epidemiology of Sarcoidosis in a Prospective Cohort Study of U.S. Women. Ann Am Thorac Soc 2016;13(1):67–71. DOI: 10.1513/AnnalsATS.201508-568BC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Newman LS, Rose CS, Maier LA. Sarcoidosis. N Engl J Med 1997;336(17):1224–34. DOI: 10.1056/NEJM199704243361706. [DOI] [PubMed] [Google Scholar]
