Open access

Genetic Factors Involved in Sarcoidosis

Written By

Birendra P. Sah and Michael C. Iannuzzi

Submitted: 19 April 2012 Published: 13 March 2013

DOI: 10.5772/55116

From the Edited Volume

Sarcoidosis

Edited by Yoshinobu Eishi

Chapter metrics overview

2,099 Chapter Downloads

View Full Metrics

1. Introduction

Sarcoidosis is an immune mediated disease thought to be caused by complex interaction between genetic and environmental factors. Involvement of genetic factors in sarcoidosis is supported by familial clustering, increased concordance in monozygotic twins and varying incidence and disease presentation among different ethnic groups. Studies have revealed several human leukocyte antigen (HLA) and non-HLA alleles consistently associated with sarcoidosis susceptibility. Two genome scans have been reported in sarcoidosis: one in African Americans reporting linkage to chromosome 5 and the other in German families reporting linkage to chromosome 6. Follow-up studies on chromosome 6 identified the BTNL2 gene, a B7 family costimulatory molecule to be associated with sarcoidosis. Recent genome-wide association studies have found annexin A11 and RAB23 genes associated with sarcoidosis. The ongoing refinement of genetic marker maps, genotyping technology, and statistical analyses makes genomic exploration for sarcoidosis genes appealing.

Advertisement

2. Evidence for genetic predisposition to sarcoidosis

Familial sarcoidosis was first noted in Germany in 1923 by Martenstein, who reported two affected sisters [1]. After that several familial cases were reported across Europe and USA. Worldwide surveys revealed that familial sarcoidosis occurred in 10.3% cases from the Netherlands [2], 7.5% from Germany [3], 5.9% from the United Kingdom [4], 4.7% from Finland [5], 4.3% from Japan [5], 9.6% from Ireland[6] and 6.9 % from Sweden[7]. A family history survey of Detroit clinic–based population in USA showed that 17% of African Americans and 3.8% of white American reported a family history in first- and second degree relatives[8]. In African Americans, the sibling recurrence risk ratio, which compares disease risk among siblings with the disease prevalence in the general population, is about 2.2 (95% confidence interval [CI], 1.03–3.68) [9].

The main limitation of these familial reports is the lack of a comparison group, and therefore it was unclear whether variation in familial sarcoidosis is due to variation in familial aggregation of disease risk, disease prevalence, or both. This question was addressed in the multicenter Case-Control Etiologic Study of Sarcoidosis (ACCESS) which evaluated 706 cases and matched controls [10]. It showed that the siblings of the affected patients had the highest relative risk (odds ratio =5.8 and 95% confidence interval=2.1–15.9). The odds ratio for the parents was 3.8 (95% CI=1.2–11.3) [10]. White cases had a markedly higher familial relative risk compared with African-American cases (18.0 versus 2.8; p=0.098).

A registry-based twin study in the Danish and the Finnish population showed an 80-fold increased risk of developing sarcoidosis in monozygotic co-twins and 7-fold increased risk in dizygotic twins [11].

Differences in disease incidence among different ethnic and racial groups exist worldwide. In the United States, African Americans have about a threefold higher age-adjusted annual incidence; 35.5 per 100,000 compared with Caucasians, 10.9 per 100,000. African American females aged 30 to 39 years were found at greatest risk at 107/100,000.The lifetime risk was calculated to be 2.4% for African Americans and 0.85% for Caucasian Americans [12]. In the United Kingdom, prevalence of sarcoidosis was found to be three times higher in the Irish living in London than in native Londoners [14]. It was eight time more common in natives of Martinique living in France than in the indigenous French populations [14]. In London the annual incidence of sarcoidosis has been reported as 1.5 per 100, 000 for Caucasians, 16.8 per 100, 000 for Asians and 19.8 per 100, 000 for Africans [15]. A study of a Swedish urban population reported a lifetime risk of 1.0% and 1.3% for men and women, respectively [16]. In addition to differences in the incidence, the clinical presentation of sarcoidosis also shows characteristic variability between ethnic groups. In both Blacks and Asians the disease has been reported to be more common, more severe and more extensive than in Caucasians [13, 15].

Advertisement

3. Genetics of other granulomatous disease

Blau syndrome and Crohn’s disease

Among the granulomatous diseases with a putative genetic component, perhaps the most intriguing are Blau syndrome and Crohn’s disease. Blau syndrome is an autosomal dominant granulomatous disease which is characterized by an early onset (before age 20) and involvement of skin, eye, and joints, similar to sarcoidosis. The factors that distinguish Blau syndrome from sarcoidosis are a lack of pulmonary involvement and absence of Kveim reactivity [17]. Crohn’s disease is a familial granulomatous inflammatory bowel disease which, like sarcoidosis, may present with uveitis, arthritis and skin rash. Crohn’s disease may involve the lung however the pattern of lung involvement differs from sarcoidosis.

Mutation in CARD (caspase activating recruitment domain) 15 gene, located on chromosome 16, is responsible for Blau syndrome [17, 18] and Crohn’s disease [19]. Nucleotide oligomerization domain protein-2 (NOD2), encoded by CARD15, recognizes peptidoglycan, a component of bacterial cell walls, and is expressed mainly by antigen-presenting cells and epithelial cells [20]. Activation of NOD2 leads to nuclear factor (NF)-кB activation [20]. Rybicki and colleagues tested 35 African American affected sib pairs by using exclusion mapping and showed that the Blau syndrome/IBD1 locus did not confer risk for sarcoidosis [21]. Schurmann and coworkers [22] evaluated four main coding CARD15 polymorphisms associated with increased risk of Crohn’s disease in both case–control and family-based sarcoidosis samples and concluded that CARD15 mutations play no role in sarcoidosis. Kanazawa and colleagues using a small sample analyzed 10 patients with early-onset sarcoidosis who had disease onset ranging from 6 months to 4 year of age and found that 9 of the 10 cases had heterozygous missense mutations in the CARD15 gene [23]. In conclusion, while an attractive candidate, no firm evidence exists to support a role for CARD 15 in sarcoidosis risk.

Chronic beryllium disease

Chronic beryllium disease (CBD), a chronic granulomatous lung disease caused by exposure to beryllium, shares similar histological and clinical findings with sarcoidsois. Glu69, carried by allele HLADPB1* 0201, was found not to be associated with sarcoidosis [24, 25]. In a study of 33 cases and 44 exposed persons without CBD (controls), Richeldi and colleagues found Glu69 in 97% of cases and in 30% of control subjects [26]. This HLA-DPB1 Glu69 association in beryllium disease has been widely supported [27] but is not associated with sarcoidosis.

Tuberculosis and leprosy

Polymorphic variants of the natural resistance–associated macrophage protein-1 gene (NRAMP1), now named SLC11A1, have been found to be associated with tuberculosis and leprosy susceptibility in endemic areas of disease [28, 29]. SLC11A1 is expressed primarily in macrophages and polymorphonuclear leukocytes and immunolocalization studies demonstrate the presence of NRAMP1 in lysosomes [30]. SLC11A1, an attractive candidate, was found not to increase the risk of sarcoidosis among African Americans [31], although a more recent article has noted an association in Polish patients (OR, 1.68; 95% CI, 1.01–2.81) [32].

Advertisement

4. Genetic associatiation studies in sarcoidosis

Genetic studies in sarcoidosis have gone through three phases – candidate gene studies, genome scanning using affected sib pair (ASP) linkage analysis and most recently, genome wide association studies (GWAS).

4.1. Candidate gene approach

The search for sarcoidosis susceptibility genes has generally relied on the candidate gene approach [33]. Investigators have selected genes for study that fit into the prevailing disease model. Sarcoidosis is thought to be a dysregulated response to an inhaled antigen that involves antigen-presenting cells, T cells (primarily a helper T-cell type 1 polar response), and cytokine and chemokine release resulting in cell recruitment and the formation of granulomas in involved organs.

4.1.1. Association with Human Leukocyte Antigens (HLA)

HLA genes have been the best studied candidate genes in sarcoidosis. HLA genes are involved in presenting antigen to T cells and are grouped into three classes: class I, II and III. HLA association studies in sarcoidosis began over thirty years ago. A summary of the most consistent HLA associations in sarcoidosis is shown in Table 1. In 1977 Brewerton and colleague [34] first revealed an association of acute sarcoidosis with the HLA class I antigen HLA-B8 which was later confirmed by other groups [35, 36]. Hedfors and co-workers [35] also noted that HLA-B8/DR3 genes were inherited as a sarcoidosis risk haplotype in whites. In white HLA-B8/DR3 haplotype is associated with wide variety of autoimmune diseases [37]. These earlier studies of class I HLA antigens directed to the studies focused on HLA class II. A recent report by Grunewald and colleagues [38] suggests that HLA class I and II genes work together in sarcoidosis pathophysiology.

HLA gene HLA class Chromosome location Risk Alleles Putative Functional Significance
HLA-A Class I 30,018, 309-
30, 021, 041 bp
A*1 Susceptibility
HLA-B Class I 31, 431, 922-
31, 432, 914 bp
B*8 Susceptibility in several populations
HLA-DQB1 Class II 32, 735, 918-
32, 742, 420 bp
*0201
*0602
Protection, Lofgren’s syndrome, mild disease in several populations
Susceptibility/disease progression in several groups
HLA-DRB1 Class II 32, 654, 526-
32, 665, 559 bp
*0301
*01, *04
*1101
Acute onset/good prognosis in several groups
Protection in several populations
Susceptibility in whites and African Americans. Stage II/III chest X-ray
HLA-DRB3 Class II 32, 654, 526-
32, 665, 540 bp
*1501

*0101
Associated with Lofgren’s syndrome
Susceptibility/disease progression in whites
BTNL2 Class II 32, 470, 490-
32, 482, 878 bp
rs2076530 BTNL2 rs2076530 G → A is associated with sarcoidosis risk in white patients but not in black patients.

Table 1.

Summary of the most consistent HLA association studies in Sarcoidosis.

Among the HLA class II antigens, HLA-DRB1 have been the most studied antigen associated with sarcoidosis. The variation in the HLA-DRB1 gene affects both susceptibility and prognosis in sarcoidosis [39, 40]. In the ACCESS study, the HLA-DRB1* 1101 allele was associated with sarcoidosis both in blacks and whites (p<0.01) and had a population attributable risk of 16% in blacks and 9% in whites [41]. In addition susceptibility markers, the ACCESS study also found that HLA class II alleles might be markers for different phenotypes of sarcoidosis such as RB1*0401 for eye involvement in blacks and whites, DRB3 for bone marrow involvement in blacks, and DPB1*0101 for hypercalcemia in whites [41]. Another consistent finding across populations has been the HLA-DQB1*0201 allele association with decreased risk and lack of disease progression [42]. Other reports strongly support the notion that several different HLA class II genes acting either in concert or independently predispose to sarcoidosis [42-44]. Linkage disequilibrium (LD) within the major histocompatibility complex (MHC) region limits the ability to precisely identify the involved HLA genes. LD exists when alleles at two distinctive loci occur in gametes more frequently than expected. Grunewald and colleagues showed that the HLA-DRB1*03 associated with resolved disease and HLA-DRB1*15 with persistent disease were synonymous with HLA-DQB1*0201 with resolved disease and HLA DQB1*0602 with persistent disease [38]. Consequently, determining the effects of HLA-DQB1 on sarcoidosis risk apart from DRB1 or dissecting out other gene effects from closely linked haplotypes in the MHC region may be an intractable problem in whites. In African Americans, HLA-DRB1/DQB1 LD may not be as strong as in Caucasians [45].

HLA alleles have been consistently associated with disease course which suggests that HLA may play greater role in determining phenotype. Furthermore, the discrepant findings in HLA association among susceptibility studies could be explained by the phenotype variation in composition of the sarcoidosis patient groups studied.

4.1.2. Association with Non-HLA candidate genes

Genes that influence antigen processing, antigen presentation, macrophage and T-cell activation, and cell recruitment and injury repair may be considered sarcoidosis candidate genes. A summary of non-HLA candidate genes reported to date is shown in Table 2.

Angiotensin-Converting Enzyme

Angiotensin-converting enzyme (ACE) is produced by sarcoidal granulomas and its serum level can be elevated in sarcoidosis. Serum ACE levels are thought to reflect granuloma burden. The ACE gene insertion (I)/deletion (D) polymorphism partially accounts for the serum ACE level variation, and investigators have proposed that this genotype should be used to adjust serum ACE reference values [46]. Studies to support a role for ACE gene polymorphisms in susceptibility or severity have been inconsistent. While only a few case control studies have suggested that ACE gene polymorphism is associated with sarcoidosis susceptibility and disease severity [47, 48], most of the studies does not support that findings [50-53].

Candidate Gene Chromosome Location Association*† Putative Functional Significance
Angiotensin-converting enzyme (ACE) 17q23 C Increased risk for ID and DD genotypes.
Moderate association between II genotype and radiographic progression.
C-C chemokine receptor 2 3p21.3 C+/- Protection/Lofgren’s syndrome association
C-C chemokine receptor 5 3p21.3 C- Association of CCR5Delta32 allele more common in patients needing corticosteroid therapy.
Refuted with haplotype analysis and larger sample.
Clara cell 10 kD protein 11q 12-13 C An allele associated with sarcoidosis and with progressive disease at 3 year follow-up.
Complement receptor 1 1q32 A The GG genotype for the Pro1827Arg
(C (5,507) G) polymorphism was significantly associated with sarcoidosis.
Cystic fibrosis trans- membrane regulator 7q31.2 A+/- R75Q increases risk.
HSPA1L heat shock protein 70 1 like 6p21.3 c HSP(+2437)CC associated with susceptibility and LS
Inhibitor kβ-α 14q13 C Association with -297T allele. Association of haplotype GTT at -881, -826, and -297, respectively. Allele -827T in Stage II.
Interleukin -1α 2q14 A The IL-1α -889 1.1 genotype increased risk.
Interleukin -4 receptor 16p11.2 No association detected in 241 members of 62 families
Interleukin -18 11q22 A+/- Genotype -607CA increased risk over AA.
No association with organ involvement.
Interferon-γ 9p22 A IFNA17 polymorphism (551T→G) and IFNA10
(60A) IFN-α 17 (551G) haplotype increased risk.
Toll-like receptor (TLR) 4
TLR10-TLR1-TLR6 cluster
9q32
4
B Asp299Gly and Thre399Ile mutations associated with chronic disease
Genetic variation in this cluster is associated with increased risk of chronic disease
Transforming growth factor (TGF) 19q13.2 B TGF-β2 59941 allele, TGF-β3 4875 A and 17369 C alleles were associated with chest X-ray detection of fibrosis.
Tumor necrosis factor
(TNF-α)
6p21.3 C+/- Genotype -307A allele associated with Lofgren’s syndrome and erythema nodosum and -857T allele with sarcoidosis. -307A not associated in African Americans.
Vascular endothelial growth factor(VEGF) 6p12 C Protective effect of +813 CT and TT genotypes.
Lower FEV1/FVC ratio observed with -627 GG genotype.
Vitamin D receptor 12q12-14 A- B allele elevated in sarcoidosis patients

Table 2.

A summary of Non-HLA candidate gene associated with Sarcoidosis

* Type of association: A = susceptibility; B = disease course; C = both.


† Association replicated (+); association refuted (-)


CC-Chemokine Receptor 2 (CCR2]

CCR 2, a receptor for monocyte chemoattractant protein, plays an important role in recruiting monocytes, T-cells, natural killer cells and dendritic cells [54]. CCR2 knockout mice die rapidly when challenged with mycobacteria [55] and display decreased IFN-γ production when challenged with Leishmania donovani or Cryptococcus neoformans [56, 57]. A single nucleotide polymorphism (SNP) in CCR2 gene (G190A, Val64Ile) is associated with protection in Japanese patients [58]. Evaluation of eight SNPs in the CCR2 gene in 304 Dutch patients showed that haplotype 2 was associated with Lofgren’s syndrome [59]. Underrepresentation of the Val64Ile variant was observed in 65 Czech patients and in 80 control subjects but did not achieve statistical significance [60]. Despite using case control–based and family-based study designs and a sample much larger than the previous three studies, Valentonyte and colleagues could not replicate the CCR2 association [61].

C-C chemokine Receptor 5 (CCR5)

CCR5 serves as a receptor for CCL3 (macrophage inflammatory protein 1-α), CCL4 (macrophage inflammatory protein 1-β), CCL5 (RANTES [regulated upon activation, T-cell expressed and secreted]), and CCL8 (monocyte chemotactic protein 2) [62, 63]. A 32 bp deletion in the CCR5 gene results in a non-functional receptor unable to bind its ligands [64]. Petrek and colleagues reported that 32-bp deletion in CCR5 gene was significantly increased in Czech patients [60], whereas Spagnolo and colleagues, using haplotype analysis, found no association in evaluating 106 white British patients and 142 control subjects and 112 Dutch patients and 169 control subjects [65].

Clara cell 10 kD protein gene

Clara cells act as stem cells in bronchial epithelial repair, provides xenobiotic metabolism, and counter regulates inflammation [66]. Clara cell 10-kD protein (CC10) has been shown to inhibit IFN-γ, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β. Murine and human CC10 gene promoter regions contain sites where inflammatory mediators, such as TNF-αand INF-α, -β, and –γ, alter transcriptional activity [67]. Increased level of CC10 in serum and BAL has been found in sarcoidosis patients whose disease had resolved compared with those whose disease had progressed [68]. The CC10 gene consists of three short exons separated by a long first and short second intron. An adenine to guanine substitution at position 38 (A38G) downstream from the transcription initiation site within the noncoding region of exon 1 has been the most studied CC10 polymorphism. The A/A genotype is believed to result in decreased CC10 levels [69]. The CC10A allele was found to be associated with sarcoidosis by Ohchi and colleague [70]. However association with the CC10 A38G polymorphism was not replicated in Dutch population or in Japanese subjects by Janssen and colleagues [71].

Complement receptor 1

Complement receptor 1 (CR1; CD35) is present on polymorphonuclear leukocytes, macrophages, B lymphocytes, some T lymphocytes, dendritic cells, and erythrocytes [71]. Immune complexes bound to CR1 are transferred to phagocytes as erythrocytes traverse the liver and spleen [72]. Immune complex clearance rates correlate with CR1 density. Low expression of erythrocyte CR1 is associated with impaired immune complex clearance and deposition outside the reticuloendothelial system [73]. These extrareticuloendothelial immune complex deposits incite local inflammatory responses and presumably granuloma formation. That immune complexes may be involved in sarcoidosis was suggested in the early 1970s. In a series involving 3,676 patients from 11 cities around the world, James and coworkers [74] reported elevated serum γ-globulin levels above 3.5 g/100 ml in 23 to 96% of patients, with IgG being the most consistently and persistently elevated [75]. The different sensitivities of the techniques used explain in part the wide range in γ-globulin levels. It is generally accepted that immune complexes are always present in sarcoidosis depending on when and how they are detected. Zorzetto and colleagues have been the only group to report a CR1 gene association with sarcoidosis [76]. The GG genotype for the Pro1827Arg (C507G) polymorphism was associated with sarcoidosis versus healthy control subjects (odds ratio [OR), 3.13; 95% CI, 1.49–6.69) and versus control subjects with chronic obstructive pulmonary disease (OR, 2.82; 95% CI, 1.27–6.39). The GG genotype was most strongly associated with disease in female patients (OR, 7.05; 95% CI, 3.10–1.61) versus healthy control subjects. No relationship with clinical variables was found.

Cystic fibrosis transmembrane conductance regulator

The R75Q mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) occurs in high frequency in patients with atypical mild cystic fibrosis [77], bronchiectasis, and allergic bronchopulmonary aspergillosis [78]. Bombieri and colleagues reported a R75Q association with sarcoidosis [79], but in followup using complete cystic fibrosis gene mutation screening they could not replicate their findings [80]. Schurmann and colleagues could not demonstrate a CFTR association with sarcoidosis [81].

Heat shock protein A1L

Heat shock proteins (HSPs) comprise a conserved group of proteins with an average weight of 70 kD. Intracellular HSPs serve as molecular chaperones [82], whereas extracellular HSPs induce cellular immune responses [83]. HSPs may also act as carrier molecules for the immunogenic peptides presented on antigen-presenting cells [84]. Polymorphisms in the HSPA1L (alias HSP70-hom) have been associated with susceptibility to rheumatoid arthritis [85]. Antibodies to HSP70 in sarcoidosis have been reported [86, 87]. To further evaluate the role of HSPs in sarcoidosis, the HSP70 +2437 C allele was evaluated and found to be associated with sarcoidosis and Lo° fgren’s syndrome in Polish patients [88] but not in Japanese patients [89].

Inhibitor κB-α

Inhibitor κB (IκB) masks the nuclear factor (NF)- κB nuclear localization sequence, thus retaining NF-κB in the cytoplasm and preventing DNA binding. On phosphorylation, IκB degrades, allowing NF-kB’s nuclear localization and initiation of transcription [90]. Terminating the NF-κB response requires IκB-α. IκB-α knockout mice die 7 to 10 days after birth with increased levels of TNF-α mRNA in the skin and severe dermatitis [91]. NF-κB–dependent signaling in alveolar macrophage makes NF-κB and thus IκB central to sarcoid pathophysiology [92]. Abdallah and colleagues found the promoter -297T allele associated with sarcoidosis [93]. No other IκB studies in sarcoidosis have been reported.

Interlukin-1(IL-1)

IL-1β produced mainly by macrophages maintains T-cell alveolitis and granuloma formation. Hunninghake and colleagues also demonstrated higher IL-1β activity in the BALF of patients with sarcoidosis compared with normal subjects [94]. Mikuniya and colleagues suggested that the ratio of IL-1 receptor antagonist to IL-1β in sarcoidal alveolar macrophage culture supernatants could predict disease chronicity [95]. The IL-1α 5’ flanking –889 C allele was found nearly two times more commonly among Czech patients with sarcoidosis compared with control subjects [96].

Interleukin Receptor- 4 (IL-4R)

The inflammatory response in sarcoidosis is primarily Th1 mediated. IL-4 drives Th2 differentiation [97]. To test whether variation in the IL-4R gene confers susceptibility to sarcoidosis, Bohnert and colleagues typed 241 members of 62 families with 136 affected siblings and 304 healthy control subjects for three functional SNPs within the IL-4R gene and found no evidence for linkage or association, thus excluding a significant role for IL-4R [98].

Interlukin-18 (IL-18)

IL-18 produced by monocytes/macrophages induces IFN-γ and drives the Th1 response. BALF and serum IL-18 levels are increased in sarcoidosis [99]. An association between IL-18607 (A/C) polymorphism and sarcoidosis has been reported and refuted in Japanese [100, 101] and white subjects [102, 103].

Interferon–α (IFN-α)

The increasing number of reported cases of IFN-α–induced sarcoidosis supports that IFN-α is important in sarcoidosis [104]. Akahoshi and colleagues found an IFN-α T551G (Ile184Arg) polymorphism associated with sarcoidosis susceptibility (OR, 3.27; 95% CI, 1.44–7.46; p=0.004) [105]. This allele is also associated with high IFN-α production and subsequent strong Th1 polarization.

Transforming Growth Factor-β (TGF-β)

Polymorphisms for all three isoforms of transforming growth factor (TGF) – β (TGF- β1, TGF-β2, and TGF-β3) have been associated with protein expression variation or functionality changes [106]. TGF-β1 levels are increased in patients with sarcoidosis who have impaired pulmonary function [107]. Kruit and colleagues reported that the TGF-β2 59941Gallele and the TGF-β3 4875 A and 17369 C alleles were associated with chest X-ray evidence of pulmonary fibrosis [85]. The TFG-β3 15101 G allele was lower in patients with fibrosis [108].

Toll-like receptor 4 (TLR4) and TLR10-TLR1-TLR6 cluster

Toll-like receptor 4 (TLR4), the first and best described of the many TLRs, plays a crucial role in detecting infection and inducing inflammatory and adaptive immune responses [109]. Pabst and colleagues examined 141 white German patients and control subjects for the TLR4 polymorphisms Asp299Gly and Thre399Ile and found no association with disease presence but did find a significant correlation with chronic disease [110].

Recently Veltcamp and colleague found that genetic variation in TLR10-TLR1-TLR6 cluster is associated with increased risk of chronic disease [111].

Tumor Necrosis Factor–α (TNF-α)

TNF-α has a broad range of inflammatory and immunostimulatory actions, including orchestrating granuloma formation. TNF-α stimulates cytokine production, enhances expression of adhesion molecules, and acts as a costimulator of T-cell activation. Alveolar macrophages from patients with active sarcoidosis secrete more TNF-α than those with inactive disease [112]. TNF-α has been considered a target for therapy in sarcoidosis [113].

Although it is unclear whether TNF-α promoter polymorphisms are functionally significant, studies suggest that a small but significant effect of the TNF-α promoter -307 A/G polymorphism may exist, with the A allele being associated with slightly greater levels of TNF-α transcription [114, 115]. A higher frequency of TNF-307A allele has been found in patients presenting with Lofgren’s syndrome and erythema nodosum [116–118]. In evaluating five promoter polymorphisms, Grutters and colleagues found a significant increase in TNF -857T allele in white British and Dutch patients and confirmed the TNF -307A allele association with Lo° fgren’s syndrome [119]. In these studies, it is not clear whether TNF-307A confers independent risk from HLA-DRB1 because TNF is in tight LD with HLA-DRBI [120]. Using a family-based approach, TNF-α was not found to be significantly associated with sarcoidosis [49].

Vascular endothelial growth factor

Dysregulated vascular endothelial growth factor (VEGF) expression has been implicated in several inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel diseases [121, 122]. VEGF modulates angiogenesis, enhances monocyte migration, a key event in granuloma formation [123]. Tolnay and colleagues reported increased VEGF transcription and protein production in activated alveolar macrophages in epithelioid cells and multinuclear giant cells of pulmonary sarcoidal granulomas [124]. Several polymorphisms have been associated with VEGF protein production [125, 126]. Morohashi and colleagues found that the VEGF+813T allele was underrepresented (associated with decreased risk) in patients with sarcoidosis. The +813 site is predicted to lie within a potential transcription factor binding site and could potentially reduce VEGF expression [126].

Vitamin D receptors

The active form of vitamin D, 1,25-dihydroxy vitamin D3, modulates the immune response through control of cytokine expression, including IFN-γ and IL-2 [127]. Increased expression of vitamin D receptors (VDRs) on sarcoidal BAL T cells and alveolar macrophage production of 1,25-dihydroxy vitamin D3 have been reported [128, 129]. Niimi and colleagues reported a VDR Bsm1 restriction site polymorphism in intron 8 to be associated with sarcoidosis [130]. Guleva and Seitzer examined a VDR Taq1 polymorphism in linkage disequilibrium with the BsmI polymorphism in 85 patients and 80 control subjects and could not confirm Niimi and colleagues’ findings [131]. Rybicki and colleagues also could not confirm VDRs as candidate genes in sarcoidosis [49].

CD80 and CD86

The B7 family of costimulatory molecules (CD80 and CD86) regulate T-cell activation. T-cell activation requires two signals: one mediated by T-cell receptor interaction with specific antigen in association with HLA molecules and an antigen-independent costimulatory signal provided by interaction between CD28 on T-cell surface and its ligands CD80 (B7-1) and CD86 (B7-2) on the antigen-presenting cells [146]. Handa and colleagues investigated CD80 and CD86 SNPs for sarcoidosis susceptibility in 146 Japanese patients and found no significant difference compared with 157 control subjects [147].

Unfortunately none of candidate gene chosen based on its likely function in sarcoidosis pathophysiology has been confirmed using the family-based study design. Limitation to many of these studies likely resides in the case-control study design’s susceptibility to a form of confounding known as population stratification which can be overcome by using a family-based design that involves recruiting patients ‘siblings and parents if available. In this design, parental alleles not transmitted to affected offspring are used as the control alleles and thus control for genetic background. The transmission disequilibrium test, one of the statistical methods used, counts the number of parental gene variants transmitted to affected offspring. Deviation from expected transmission supports a predisposing effect of the more frequently transmitted allele.

4.2. Genome scanning: Affected sib pair linkage analysis

Sarcoidosis genome scan in Germans

The first genome scan study related to sarcoidosis was conducted by Schurmann and colleagues, in which they used 225 microsatellite markers spanning the genome in 63 German families to identify a linkage signal (D6S1666) on chromosome 6p21 [132]. This group then used a three-stage single-nucleotide polymorphism (SNP) scan of the 16-MB region surrounding D6S1666 [133] and identified a single SNP, rs2076530, in the BTNL2 gene associated with sarcoidosis. This SNP (G/A) was found at the 3’ boundary of the exon 5 coding region. The A allele at this position has been proposed to introduce an alternative splice site at the exon 5–3’ intron boundary of the BTNL2 transcript that results in a premature truncation of the protein.

BTNL2, also known as “butyrophilin-like 2” and “BTL-2,” is a butyrophilin gene that belongs to the immunoglobulin gene superfamily related to the B7 family [134, 135]. Butyrophilin was initially cloned from cattle mammary epithelial cells [136]. This gene was localized to the MHC class II region in humans. To determine the consistency of the BTNL2 gene as a sarcoidosis risk factor across different populations, Rybicki and colleagues characterized variation in the BTNL2 exon/intron 5 region in an African-American family sample that consisted of 219 nuclear families (686 individuals) and in 2 case–control samples (295 African-American matched pairs and 366 white American matched pairs) [137].They confirmed that BTNL2 somewhat was less associated with sarcoidosis in African Americans compared with whites. BTNL2 appears to have moderate influence on individual disease risk (odds ratio of 1.6 in heterozygotes and 2.8 in homozygotes). The population attributable risk of 23% for heterozygotes and homozygotes indicates a significant contribution at the population level.

Whether BTNL2 as a sarcoidosis risk factor is independent of HLA-DRB risk alleles or not, still remains a question. HLADRB and BTNL2 are in linkage disequilibrium. Linkage disequilibrium is the nonrandom association of alleles physically closes on a chromosome. HLA-DRB lies about 180 kb centromeric to BTNL2. On the basis of regression models, BTNL2 appears to be an independent risk factor [133, 137]. In the case of blacks, in whom the BTNL2-conferred sarcoidosis risk is less significant than for whites, a negative interaction with HLA-DR appears to exist [137]. In one study, BTNL2 was found not to be associated with Wegener’s granulomatosis [138].

Most recently Hofmann and colleagues [139] conducted a Genome-Wide Linkage Analysis in 181 German sarcoidosis families using clustered biallelic markers. This study revealed one region of suggestive linkage on chromosome 12p13.31 at 20 cM (LOD= 2.53; local P value =.0003) and another linkage on 9q33.1 at 134 cM (LOD =2.12; local P value =.0009). It is proposed that these regions might harbor yet-unidentified, possibly subphenotype-specific risk factors for the disease (e.g. immune-related functions like the tumor necrosis factor receptor 1).

Sarcoidosis genome scan in African Americans

Eleven centers joined together in an NHLBI-sponsored effort (Sarcoidosis Genetic Analysis Consortium [SAGA]) to perform a genome scan in African American siblings. This group performed a 380-microsatellite genomewide scan across 22 autosomes in 519 African American sib pairs. The significant findings included 15 markers with p values < 0.05 with the strongest linkage signal on chromosome 5 [140]. Fine mapping studies indicated a sarcoidosis susceptibility gene on chromosome 5q11.2 and a gene protective effect for sarcoidosis on 5p15.2 [141].

The reason why African Americans were chosen to uncover sarcoidosis susceptibility genes was that African Americans are more commonly and severely affected and have affected family members more often than whites. But the disadvantage of doing so is that African Americans are admixed with white and other populations to varying degrees with possible admixture among their participating centers ranging from 12% in South Carolina to 20% in New York [142]. To address the possibility that admixed subpopulations existed in the SAGA sample and affected the power to detect linkage, the sample was stratified by genetically determined ancestry using the data from the 380 microsatellite markers genotyped in the genome scan. The African-American families were clustered into subpopulations based on ancestry similarity. Evidence of two genetically distinct groups was found: Stratified linkage results suggest that one subpopulation of families contributed to previously identified linkage signals at 1p22, 3p21-14, 11p15, and 17q21 and that a second subpopulation of families contributed to those found at 5p15-13 and 20q13 [143]. These findings support the presence of sarcoidosis susceptibility genes in regions previously identified but indicate that these genes are likely to be specific to ancestral groups that have combined to form modern-day African Americans.

4.3. Genome-Wide Association Studies (GWAS)

In genome-wide association study high throughput genotyping methods are used to genotype a dense set of SNPs across the genome. A significant advantage of this approach is that association studies are more powerful than affected sib pair methods of linkage analysis. Hofmann and colleagues [144] conducted a genomewide association study of 499 German patients with sarcoidosis and 490 control subjects. The strongest signal mapped to the annexin A11 gene on chromosome 10q22.3. Validation in an independent sample confirmed the association. Annexin A11 has regulatory functions in calcium signaling, cell division, vesicle trafficking, and apoptosis. Depletion or dysfunction of annexin A11 may affect the apoptosis pathway in sarcoidosis. Later the same group [145] reported another associated locus 6p12.1 that comprises several genes, a likely candidate being RAB23. RAB23 is proposed to be involved in antibacterial defense processes and regulation of the sonic hedgehog signaling pathway.

Advertisement

5. Counseling and screening

In the context of genetic family counseling, this generally is perceived as a small risk by the clients and should lead to enhanced awareness but does not justify specific medical investigations in the absence of complaints.

Advertisement

6. Genetic testing

Genetic testing at present does not play a role in the diagnosis and treatment of sarcoidosis.

Advertisement

7. Future directions

The cause of sarcoidosis remains unknown. It is thought to be caused by interaction between environmental and genetic factors. Genetic studies have revealed the HLA and other candidate genes associated with sarcoidosis susceptibility. Association studies have been motivated by the hopes that identifying alleles that affect risk and phenotype will help in understanding disease etiology. Unfortunately, many of the reported associations have not been replicated. Two genome scans have been reported and one has yielded a likely candidate gene, BTNL2 that has been replicated in large studies. Emerging technologies and advances in genomics and proteomics will help find the causes sarcoidosis, better understanding of pathogenesis of sarcoidosis and to test new therapy. Gene expression profiling in BALF and blood carried out at the time of presentation will likely help to better predict disease resolution or progression.

References

  1. 1. Martenstein H Knochveranderungen bei lupus pernioZ Haut Geschlechtskr 7308, 1923
  2. 2. WirnsbergerR. MDe VriesJWoutersE. FDrentMClinical presentation of sarcoidosis in The Netherlands an epidemiological study. Neth J Med. 1998Aug; 5325360
  3. 3. KristenDSarcoidosis in Germany. Analysis of a questionnaire survey in 1992 of patients of the German Sarcoidosis Group. Pneumologie. 1995Jun;49637882
  4. 4. McgrathD. SDaniilZFoleyPdu Bois JL, Lympany PA, Cullinan P, du Bois RM. Epidemiology of familial sarcoidosis in the UK. Thorax. 2000Sep; 5597514
  5. 5. PietinalhoAOhmichiMHirasawaMHiragaYLofroosA. BSelroosOFamilial sarcoidosis in Finland and Hokkaido, Japan-a comparative study. Respir Med 199993408412
  6. 6. BrennanN. JCreanPLongJ. PFitzgeraldM. XHigh prevalence of familial sarcoidosis in an Irish population. Thorax 1984391418
  7. 7. WimanL. GFamilial occurrence of sarcoidosis. Scand J Respir Dis Suppl 197280115119
  8. 8. HarringtonDMajorMRybickiBPopovich J Jr, Maliarik M, Iannuzzi MC. Familial analysis of 91 families. Sarcoidosis 199411240243
  9. 9. RybickiB. AKirkeyK. LMajorMMaliarikM. JPopovich J Jr, Chase GA, IannuzziMC. Familial risk ratio of sarcoidosis inAfrican-American sibs and parents. Am J Epidemiol 2001153188193
  10. 10. RybickiB. AIannuzziM. CFrederickM. MThompsonB. WRossmanM. DBresnitzE. ATerrinM. LMollerD. RBarnardJBaughmanR. Pet alFamilial aggregation of sarcoidosis: A Case-Control Etiologic Study of Sarcoidosis (ACCESS). Am J Respir Crit Care Med 200116420852091
  11. 11. SverrildABackerVKyvikK. OKaprioJMilmanNSvendsenC. BThomsenS. FHeredity in sarcoidosis: a registry-based twin study. Thorax. 2008Oct; 63108946Epub 2008 Jun 5.
  12. 12. RybickiB. AMajorMPopovich J Jr, Maliarik MJ, Iannuzzi MC. Racial differences in sarcoidosis incidence: a 5-year study in a health maintenance organization. Am J Epidemiol 1997145234241
  13. 13. RybickiB. AMaliarikM. JMajorMPopovich J Jr, Iannuzzi MC. Epidemiology, demographics, and genetics of sarcoidosis. Sem Respir Infect 19983166173
  14. 14. JamesD. GNevilleESiltzbachL. EA worldwide review of sarcoidosis. Ann N Y Acad Sci. 197627832134
  15. 15. EdmondstoneW. MWilsonA. GSarcoidosis in Caucasians, Blacks and Asians in London. Br J Dis Chest. 1985Jan; 7912736
  16. 16. HillerdalGNou EOstermanKet alSarcoidosis: epidemiology and prognosis. A 15-year European study. Am Rev Respir Dis 19841302932
  17. 17. Blau EB Familial granulomatous arthritisiritis, and rash. J Pediatr 107689493,1985
  18. 18. Miceli-richardCLesageSRybojadMPrieurA. MManouvrier-hanuSHafnerRChamaillardMZoualiHThomasGHugotJ. PCARD15 mutations in Blau syndrome. Nat Genet 2001291920
  19. 19. HugotJ. PChamaillardMZoualiHLesageSCezardJ. PBelaicheJAlmerSTyskCOMorainC. AGassullM, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease.Nature 2001411599603
  20. 20. StroberWMurrayP. JKitaniAWatanabeTSignalling pathways and molecular interactions of NOD1 and NOD2. Nat Rev Immunol 20066920
  21. 21. RybickiB. AMaliarikM. JBockC. HElstonR. CBaughmanR. PKimaniA. PShefferR. GChenK. MMajorMPopovichJJr, et alThe Blausyndrome gene is not a major risk factor for sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 199916203208
  22. 22. SchurmannMValentonyteRHampeJMuller-quernheimJSchwingerESchreiberSCARD15 gene mutations in sarcoidosis. Eur Respir J 200322748754
  23. 23. KanazawaNOkafujiIKambeNNishikomoriRNakata-hizumeMNagaiSFujiAYuasaTMankiASakuraiYet alEarly-onset sarcoidosis and CARD15 mutations with constitutive nuclear factorkappaB activation: common genetic etiology with Blau syndrome. Blood 200510511951197
  24. 24. MaliarikM. JChenK. MMajorM. LShefferR. GPopovich J Jr, Rybicki BA, Iannuzzi MC. Analysis of HLA-DPB1 polymorphisms in African-Americans with sarcoidosis. Am J Respir Crit Care Med 1998158111114
  25. 25. SchurmannMBeinGKirstenDSchlaakMMuller-quernheimJSchwingerEHLA-DQB1 and HLA-DPB1 genotypes in familial sarcoidosis. Respir Med 199892649652
  26. 26. RicheldiLSorrentinoRSaltiniCHLA-DPB1 glutamate 69: a genetic marker of beryllium disease. Science 1993262242244
  27. 27. AmicosanteMSanaricoNBerrettaFArroyoJLombardiGLechlerRColizziVSaltiniCBeryllium binding to HLA-DP molecule carrying the marker of susceptibility to berylliosis glutamate beta 69. Hum Immunol 200162686693
  28. 28. BellamyRIdentifying genetic susceptibility factors for tuberculosis in Africans: a combined approach using a candidate gene study and a genome-wide screen. Clin Sci (Lond) 200098245250
  29. 29. AbelLSanchezF. OObertiJThucN. VHoaL. VLapV. DSkameneELagrangeP. HSchurrESusceptibility to leprosy is linked to the human NRAMP1 gene. J Infect Dis 1998177133145
  30. 30. GruenheidSPinnerEDesjardinsMGrosPNatural resistance to infection with intracellular pathogens: the Nramp1 protein is recruited to the membrane of the phagosome. J Exp Med 1997185717730
  31. 31. MaliarikM. JChenK. MShefferR. GRybickiB. AMajorM. LPopovich J Jr, Iannuzzi MC. The natural resistance-associated macrophage protein gene in African Americans with sarcoidosis. Am J Respir Cell Mol Biol 200022672675
  32. 32. DubaniewiczAJamiesonS. EDubaniewicz-wybieralskaMFakiolaMNancy Miller E, Blackwell JM. Association between SLC11A1 (formerly NRAMP1) and the risk of sarcoidosis in Poland. Eur J Hum Genet 200513829834
  33. 33. IannuzziM. CRybickiB. AMaliarikMPopovich J Jr. Finding disease genes: from cystic fibrosis to sarcoidosis [Thomas A. Neff Lecture].Chest 1997S-73S
  34. 34. BrewertonD. ACockburnCJamesD. CJamesD. GNevilleEHLA antigens in sarcoidosis. Clin Exp Immunol 197727227229
  35. 35. HedforsELindstromFHla-bD. Rin sarcoidosis: correlation to acute onset disease with arthritis. Tissue Antigens 198322200203
  36. 36. SmithM. JTurtonC. WMitchellD. NTurner-warwickMMorrisL. MLawlerS. DAssociation of HLA B8 with spontaneous resolution in sarcoidosis. Thorax 198136296298
  37. 37. LioDCandoreGRomanoG. CDAnnaCGervasiFDiLorenzoGModicaM. APotestioMCaruso C. Modification of cytokine patterns in subjects bearing the HLA-B8,DR3 phenotype: implications for autoimmunity. Cytokines Cell Mol Ther 19973217224
  38. 38. GrunewaldJEklundAOlerupOHuman leukocyte antigen class I alleles and the disease course in sarcoidosis patients. Am J Respir Crit Care Med 2004169696702
  39. 39. RossmanM. DThompsonBFrederickMMaliarikMIannuzziM. CRybickiB. APandeyJ. PNewmanL. SMagiraEBeznik-cizmanBet alHLA-DRB1*1101: a significant risk factor for sarcoidosis in blacks and whites. Am J Hum Genet 200373720735
  40. 40. IshiharaMOhnoSIshidaTAndoHNaruseTNoseYInokoHMolecular genetic studies ofHLA class II alleles in sarcoidosis. Tissue Antigens 199443238241
  41. 41. RossmanM. DThompsonBFrederickMMaliarikMIannuzziM. CRybickiB. Aet alHLA-DRB1*1101: a significant risk factor for sarcoidosis in blacks and whites. Am J Hum Genet 2003734720735
  42. 42. IannuzziM. CMaliarikM. JPoissonL. MRybickiB. ASarcoidosis susceptibility and resistance HLA-DQB1 alleles in African Americans. Am J Respir Crit Care Med 200316712251231
  43. 43. MaliarikM. JChenK. MMajorM. LShefferR. GPopovich J Jr, Rybicki BA, Iannuzzi MC. Analysis of HLA-DPB1 polymorphisms in African-Americans with sarcoidosis. Am J Respir Crit Care Med 1998158111114
  44. 44. 17. RybickiB. AMaliarikM. JPoissonL. MShefferRChenK. MMajorMChaseG. AIannuzziM. CThe major histocompatibility complex gene region and sarcoidosis susceptibility in African Americans. Am J Respir Crit Care Med 2003167444449
  45. 45. ZacharyA. ABiasW. BJohnsonARoseS. MLeffellM. SAntigen, allele, and haplotype frequencies report of the ASHI minority antigens workshops: part 1, African-Americans. Hum Immunol 20016211271136
  46. 46. SharmaPSmithIMaguireGStewartSShneersonJBrownM. JClinical value of ACE genotyping in diagnosis of sarcoidosis. Lancet 199734916021603
  47. 47. MaliarikM. JRybickiB. AMalvitzEShefferR. GMajorMPopovich J Jr, Iannuzzi MC. Angiotensin-converting enzyme gene polymorphism and risk of sarcoidosis. Am J Respir Crit Care Med 199815815661570
  48. 48. PietinalhoAFuruyaKYamaguchiEKawakamiYSelroosOThe angiotensin converting enzyme DD gene is associated with poor prognosis in Finnish sarcoidosis patients.Eur Resp J 1999137236
  49. 49. RybickiB. AMaliarikM. JPoissonL. MIannuzziM. CSarcoidosis and granuloma genes: a family-based study in African-Americans. Eur Respir J 200424251257
  50. 50. PlanckAEklundAYamaguchiEGrunewaldJAngiotensin-converting enzyme gene polymorphism in relation to HLA-DR in sarcoidosis. J Intern Med. 2002Mar; 251321722
  51. 51. McgrathD. SFoleyP. JPetrekMIzakovicova-hollaLDu Bois RM. Ace gene I/D polymorphism and sarcoidosis pulmonary disease severity. Am J Respir Crit Care Med. 2001Jul 15; 1642197201
  52. 52. SchürmannMReichelPMüller-myhsokBSchwingerEAngiotensin-converting enzyme (ACE) gene polymorphisms and familial occurrence of sarcoidosis. J Intern Med. 2001Jan; 24917783
  53. 53. ArbustiniEGrassoMLeoGTinelliCFasaniRDiegoliMBanchieriNCiprianiAGorriniMSemenzatoGet alPolymorphism of angiotensin-converting enzyme gene in sarcoidosis. Am J Respir Crit Care Med 1996153851854
  54. 54. BoringLGoslingJChensueS. WKunkelS. LFarese RV Jr, Broxmeyer HE, Charo IF. Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 199710025522561
  55. 55. PetersWScottH. MChambersH. FFlynnJ. LCharoI. FErnstJ. DChemokine receptor 2 serves an early and essential role in resistance to Mycobacterium tuberculosis. Proc Natl Acad Sci USA 20019879587963
  56. 56. TraynorT. RKuzielW. AToewsG. BHuffnagleG. BCCR2 expression determines T1 versus T2 polarization during pulmonary Cryptococcus neoformans infection. J Immunol 200016420212027
  57. 57. SatoNKuzielW. AMelbyP. CReddickR. LKosteckiVZhaoWMaedaNAhujaS. KAhujaS. SDefects in the generation of IFN gamma are overcome to control infection with Leishmania donovani in CC chemokine receptor (CCR) 5-, macrophage inflammatory protein-1 alpha-, or CCR2-deficient mice. J Immunol 199916355195525
  58. 58. HizawaNYamaguchiEFuruyaKJinushiEItoAKawakamiYThe role of the C-C chemokine receptor 2 gene polymorphism 64ICCR2- 64I) in sarcoidosis in a Japanese population. Am J Respir Crit Care Med 1999
  59. 59. SpagnoloPRenzoniE. AWellsA. USatoHGruttersJ. CSestiniPAbdallahAGramiccioniERuvenH. Jdu Bois RM, et al. C-C chemokine receptor 2 and sarcoidosis: association with Lofgren’s syndrome. Am J Respir Crit Care Med 200316811621166
  60. 60. PetrekMDrabekJKolekVZlamalJWelshK. IBunceMWeiglEDu Bois R. CC chemokine receptor gene polymorphisms in Czech patients with pulmonary sarcoidosis. Am J Respir Crit Care Med 200016210001003
  61. 61. ValentonyteRHampeJCroucherP. JMuller-quernheimJSchwingerESchreiberSSchurmannMStudy of C-C chemokine receptor 2 alleles in sarcoidosis, with emphasis on family-based analysis. Am J Respir Crit Care Med 200517111361141
  62. 62. BlanpainCMigeotteILeeBVakiliJDoranzB. JGovaertsCVassartGDomsR. WParmentierMCCR5 binds multiple CC-chemokines: MCP-3 acts as a natural antagonist. Blood 19999418991905
  63. 63. CombadiereCAhujaS. KTiffanyH. LMurphyP. MCloning and functional expression of CC CKR5, a human monocyte CC chemokine receptor selective for MIP-1(alpha), MIP-1(beta), and RANTES. J Leukoc Biol 199660147152
  64. 64. MantovaniAThe chemokine system: redundancy for robust outputs. Immunol Today. 1999Jun;2062547
  65. 65. SpagnoloPRenzoniE. AWellsA. UCopleyS. JDesaiS. RSatoHGruttersJ. CAbdallahATaegtmeyerAdu Bois RM, et al. C-C chemokine receptor 5 gene variants in relation to lung disease in sarcoidosis. Am J Respir Crit Care Med 2005172721728
  66. 66. SinghGKatyalS. LClara cells and Clara cell 10 kD protein (CC10). Am J Respir Cell Mol Biol 199717141143
  67. 67. CowanM. JHuangXYaoX. LShelhamerJ. HTumor necrosis factor alpha stimulation of human Clara cell secretory protein production by human airway epithelial cells. Ann NY Acad Sci 2000923193201
  68. 68. ShijuboNItohYShigeharaKYamaguchiTItohKShibuyaYTakahashiROhchiTOhmichiMHiragaYet alAssociation of Clara cell 10-kDa protein, spontaneous regression and sarcoidosis. Eur Respir J 200016414419
  69. 69. LaingI. AHermansCBernardABurtonP. RGoldblattJLe Souef PN. Association between plasma CC16 levels, the A38G polymorphism, and asthma. Am J Respir Crit Care Med 2000161124127
  70. 70. OhchiTShijuboNKawabataIIchimiyaSInomataSYamaguchiAUmemoriYItohYAbeSHiragaYet alPolymorphism of Clara cell 10-kD protein gene of sarcoidosis. Am J Respir Crit Care Med 2004169180186
  71. 71. JanssenRSatoHGruttersJ. CRuvenH. Jdu Bois RM, Matsuura R, Yamazaki M, Kunimaru S, Izumi T, Welsh KI, et al. The Clara cell10 adenine38guanine polymorphism and sarcoidosis susceptibility in Dutch and Japanese subjects. Am J Respir Crit Care Med 200417011851187
  72. 72. LoegeringD. JBlumenstockF. ADepressing hepatic macrophage complement receptor function causes increased susceptibility to endotoxemia and infection. Infect Immun 198547659664
  73. 73. CornacoffJ. BHebertL. ASmeadW. LVanamanM. EBirminghamD. JWaxmanF. JPrimate erythrocyte-immune complex-clearing mechanism. J Clin Invest 198371236247
  74. 74. SchifferliJ. ANgY. CEstreicherJWalportM. JThe clearance of tetanus toxoid/anti-tetanus toxoid immune complexes from the circulation of humans: complement- and erythrocyte complement receptor 1- dependent mechanisms. J Immunol 1988140899904
  75. 75. JamesD. GNevilleEWalkerAImmunology of sarcoidosis. Am JMed 197559388394
  76. 76. ZorzettoMBombieriCFerrarottiIMedagliaSAgostiniCTinelliCMalerbaGCarrabinoNBerettaACasaliLet alComplement receptor 1 gene polymorphisms in sarcoidosis. Am J Respir Cell Mol Biol 2002271723
  77. 77. HughesDDorkTStuhrmannMGrahamCMutation and haplotype analysis of the CFTR gene in atypically mild cystic fibrosis patients from Northern Ireland. J Med Genet 200138136139
  78. 78. LuisettiMPignattiP. FGenetics of idiopathic disseminated bronchiectasis. Semin Respir Crit Care Med 200324179184
  79. 79. BombieriCLuisettiMBelpinatiFZulianiEBerettaABaccheschiJCasaliLPignattiP. FIncreased frequency of CFTR gene mutations in sarcoidosis: a case/control association study. Eur J Hum Genet 20008717720
  80. 80. BombieriCBelpinatiFPignattiP. FLuisettiMComment on ‘CFTR gene mutations in sarcoidosis’. Eur J Hum Genet 200311553554
  81. 81. SchurmannMAlbrechtMSchwingerEStuhrmannMCFTR gene mutations in sarcoidosis. Eur J Hum Genet 200210729732
  82. 82. MatouschekAProtein unfolding: an important process in vivo? Curr Opin Struct Biol 20031398109
  83. 83. AseaAKraeftS. KKurt-jonesE. AStevensonM. AChenL. BFinbergR. WKooG. CCalderwoodS. KHSP70 stimulates cytokine production through a CD14-dependant pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med 20006435442
  84. 84. SrivastavaP. KUdonoHBlachereN. ELiZHeat shock proteins transfer peptides during antigen processing and CTL priming. Immunogenetics 1994399398
  85. 85. JenkinsS. CMarchR. ECampbellR. DMilnerC. MA novel variant of the MHC-linked hsp70, hsp70-hom, is associated with rheumatoid arthritis. Tissue Antigens 2000563844
  86. 86. De SmetM. DRamadanACirculating antibodies to inducible heat shock protein 70 in patients with uveitis. Ocul Immunol Inflamm 200198592
  87. 87. HrycajPWurmKMennetPMullerWAntibodies to heat shock proteins in patients with pulmonary sarcoidosis. Sarcoidosis 199512124130
  88. 88. Bogunia-kubikKKoscinskaKSuchnickiKLangeAHSP70-hom gene single nucleotide (-2763 G/A and-2437 C/T) polymorphisms in sarcoidosis. Int J Immunogenet 200633135140
  89. 89. IshiharaMOhnoSIshidaTMizukiNAndoHNaruseTIshiharaHInokoHGenetic polymorphisms of the TNFB and HSP70 genes located in the human major histocompatibility complex in sarcoidosis. Tissue Antigens 1995465962
  90. 90. PerkinsN. DThe Rel/NF-kappa B family: friend and foe. Trends Biochem Sci 200025434440
  91. 91. KlementJ. FRiceN. RCarB. DAbbondanzoS. JPowersG. DBhattP. HChenC. HRosenC. AStewartC. LIkappaBalpha deficiency results in a sustained NF-kappaB response and severe widespread dermatitis in mice. Mol Cell Biol 19961623412349
  92. 92. ConronMBondesonJPantelidisPBeynonH. LFeldmannMduBois RM, Foxwell BM. Alveolar macrophages and T cells from sarcoid,but not normal lung, are permissive to adenovirus infection and allow analysis of NF-kappa b-dependent signaling pathways. Am J Respir Cell Mol Biol 200125141149
  93. 93. AbdallahASatoHGruttersJ. CVeeraraghavanSLympanyP. ARuvenH. Jvan den Bosch JM, Wells AU, du Bois RM, Welsh KI. Inhibitor kappa B-alpha (IkappaB-alpha) promoter polymorphisms in UK and Dutch sarcoidosis. Genes Immun 20034450454
  94. 94. HunninghakeG. WRelease of interleukin-1 by alveolar macrophages of patients with active pulmonary sarcoidosis. Am Rev Respir Dis 1984129569572
  95. 95. MikuniyaTNagaiSTakeuchiMMioTHoshinoYMikiHShigematsuMHamadaKIzumiTSignificance of the interleukin-1 receptor antagonist/interleukin-1 beta ratio as a prognostic factor in patients with pulmonary sarcoidosis. Respiration (Herrlisheim) 200067389396
  96. 96. HutyrovaBPantelidisPDrabekJZurkovaMKolekVLenhartKWelshK. IDu Bois RM, Petrek M. Interleukin-1 gene cluster polymorphisms in sarcoidosis and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2002165148151
  97. 97. RengarajanJSzaboS. JGlimcherL. HTranscriptional regulation of Th1/Th2 polarization. Immunol Today 200021479483
  98. 98. BohnertASchurmannMHartungAHacksteinHMuller-quernheimJBeinGNo linkage of the interleukin-4 receptor locus on chromosome 1611with sarcoidosis in German multiplex families. Eur J Immunogenet 2002
  99. 99. ShigeharaKShijuboNOhmichiMYamadaGTakahashiROkamuraHKurimotoMHiragaYTatsunoTAbeSet alIncreased levels of interleukin-18 in patients with pulmonary sarcoidosis. Am J Respir Crit Care Med 200016219791982
  100. 100. ZhouYYamaguchiEHizawaNNishimuraMRoles of functional polymorphisms in the interleukin-18 gene promoter in sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 200522105113
  101. 101. TakadaTSuzukiEMorohashiKGejyoFAssociation of single nucleotide polymorphisms in the IL-18 gene with sarcoidosis in a Japanese population. Tissue Antigens 2002603642
  102. 102. KellyD. MGreeneC. MMeacheryGOMahonyMGallagherP. MTaggartC. C, ONeillS. JMcelvaneyNG. Endotoxin up-regulates interleukin-18: potential role for gram-negative colonization in sarcoidosis. Am J Respir Crit Care Med 200517212991307
  103. 103. JanssenRGruttersJ. CRuvenH. JZanenPSatoHWelshK. Idu Bois RM, van den Bosch JM. No association between interleukin-18 gene polymorphisms and haplotypes in Dutch sarcoidosis patients. Tissue Antigens 200463578583
  104. 104. GoldbergH. JFiedlerDWebbAJagirdarJHoyumpaA. MPetersJSarcoidosis after treatment with interferon-alpha: A case series and review of the literature. Respir Med 200610020632068
  105. 105. AkahoshiMIshiharaMRemusNUnoKMiyakeKHirotaTNakashimaKMatsudaAKandaMEnomotoTet alAssociation between IFNA genotype and the risk of sarcoidosis. HumGenet 2004114503509
  106. 106. AwadM. REl-GamelAHasletonPTurnerD. MSinnottP. JHutchinsonI. VGenotypic variation in the transforming growth factor-beta1 gene: association with transforming growth factor-beta1 production, fibrotic lung disease, and graft fibrosis after lung transplantation. Transplantation 19986610141020
  107. 107. SalezFGossetPCopinM. CLamblin Degros C, Tonnel AB, Wallaert B. Transforming growth factor-beta1 in sarcoidosis. Eur Respir J 199812913919
  108. 108. KruitAGruttersJ. CRuvenH. JVan MoorselC. HWeiskirchenRMengsteabSvan den Bosch JM. Transforming growth factor-beta gene polymorphisms in sarcoidosis patients with and without fibrosis. Chest 200612915841591
  109. 109. AkiraSTakedaKKaishoTToll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol 20012675680
  110. 110. Pabst S, Baumgarten G, Stremmel A, LennarzM,Knufermann P,Gillissen A, Vetter H, Grohe C. Toll-like receptor (TLR) 4 polymorphisms are associated with a chronic course of sarcoidosis. Clin Exp Immunol 2006; 143:420-426.
  111. 111. VeltkampMVan MoorselC. HRijkersG. TRuvenH. JGruttersJ. Cet alGenetic variation in the Toll-like receptor gene cluster (TLR10-TLR1-TLR6) influences disease course in sarcoidosis. Tissue Antigens. 2012Jan; 7912532
  112. 112. ZhengLTeschlerHGuzmanJHubnerKStrizICostabelUAlveolar macrophage TNF-alpha release and BALcell phenotypes in sarcoidosis. Am J Respir Crit Care Med 199515210611066
  113. 113. BaughmanR. PIannuzziMTumour necrosis factor in sarcoidosis and its potential for targeted therapy. BioDrugs 200317425431
  114. 114. AllenR. DPolymorphism of the human TNF-alpha promoter: random variation or functional diversity? Mol Immunol 19993610171027
  115. 115. WilsonA. Gdi Giovine FS, Blakemore AI, Duff GW. Single base polymorphism in the human tumour necrosis factor alpha (TNF alpha) gene detectable by NcoI restriction of PCR product. Hum Mol Genet 1992
  116. 116. SomoskoviAZisselGSeitzerUGerdesJSchlaakMMuller Quernheim J. Polymorphisms at position-308 in the promoter region of the TNF-alpha and in the first intron of the TNF-beta genes and spontaneous and lipopolysaccharide-induced TNF alpha release in sarcoidosis. Cytokine 199911882887
  117. 117. SeitzerUSwiderCStuberFSuchnickiKLangeARichterEZabelPMuller-quernheimJFladH. DGerdesJTumour necrosis factor alpha promoter gene polymorphism in sarcoidosis. Cytokine 19979787790
  118. 118. LabunskiSPosernGLudwigSKundtGBrockerE. BKunzMTumour necrosis factor-alpha promoter polymorphism in erythema nodosum. Acta Derm Venereol 2001811821
  119. 119. GruttersJ. CSatoHPantelidisPLaganA. LMcgrathD. SLammersJ. Wvan den Bosch JM,Wells AU, du Bois RM, Welsh KI. Increased frequency of the uncommon tumor necrosis factor-857T allele in British and Dutch patients with sarcoidosis. Am J Respir Crit Care Med 200216511191124
  120. 120. WilsonA. GDe VriesNPociotFdi Giovine FS, van der Putte LB, Duff GW. An allelic polymorphism within the human tumor necrosis factor alpha promoter region is strongly associated with HLA A1, B8, and DR3 alleles. J Exp Med 1993177557560
  121. 121. KanazawaSTsunodaTOnumaEMajimaTKagiyamaMKikuchiKVEGF, basic-FGF, and TGF-beta in Crohn’s disease and ulcerative colitis: a novel mechanism of chronic intestinal inflammation. Am J Gastroenterol 200196822828
  122. 122. KasamaTShiozawaFKobayashiKYajimaNHanyudaMTakeuchiH. TMoriYNegishiMIdeHAdachiMVascular endothelial growth factor expression by activated synovial leukocytes in rheumatoid arthritis: critical involvement of the interaction with synovial fibroblasts. Arthritis Rheum 20014425122524
  123. 123. FlammeIFrolichTRisauWMolecular mechanisms of vasculogenesis and embryonic angiogenesis. J Cell Physiol 1997173206210
  124. 124. TolnayEKuhnenCVossBWiethegeTMullerK. MExpression and localization of vascular endothelial growth factor and its receptor flt in pulmonary sarcoidosis. Virchows Arch 19984326165
  125. 125. WatsonC. JWebbN. JBottomleyM. JBrenchleyP. EIdentification of polymorphisms within the vascular endothelial growth factor (VEGF) gene: correlation with variation in VEGF protein production. Cytokine 20001212321235
  126. 126. RennerWKotschanSHoffmannCObermayer-pietschBPilgerEA CommonCT mutation in the gene for vascular endothelial growth factor is associated with vascular endothelial growth factor plasma levels. J Vasc Res 200037443448
  127. 127. HewisonMVitaminDand the immune system. J Endocrinol 1992132173175
  128. 128. Biyoudi-vouenzeRCadranelJValeyreDMilleronBHanceA. JSolerPExpression of 1,25(OH)2D3 receptors on alveolar lymphocytes from patients with pulmonary granulomatous diseases. Am Rev Respir Dis 199114313761380
  129. 129. AdamsJ. SSingerF. RGacadM. ASharmaO. PHayesM. JVourosPHolickM. FIsolation and structural identification of 1, 25-dihydroxyvitamin D3 produced by cultured alveolar macrophages in sarcoidosis. J Clin Endocrinol Metab 198560960966
  130. 130. NiimiTTomitaHSatoSKawaguchiHAkitaKMaedaHSugiuraYUedaRVitamin D receptor gene polymorphism in patients with sarcoidosis. Am J Respir Crit Care Med 199916011071109
  131. 131. GulevaISeitzerUVitamin D receptor gene polymorphism in patients with sarcoidosis. Am J Respir Crit Care Med 2000162760761
  132. 132. SchurmannMReichelPMuller-myhsokBSchlaakMMuller- QuernheimJSchwingerEResults from a genome-wide search for predisposing genes in sarcoidosis. Am J Respir Crit Care Med 2001164840846
  133. 133. ValentonyteRHampeJHuseKRosenstielPAlbrechtMStenzelANagyMGaedeK. IFrankeAHaeslerRet alSarcoidosis is associated with a truncating splice site mutation in BTNL2. Nat Genet 200537357364
  134. 134. RhodesD. AStammersMMalcherekGBeckSTrowsdaleJThe cluster of BTN genes in the extended major histocompatibility complex. Genomics 200171351362
  135. 135. SharpeA. HFreemanG. JTheBCD28 superfamily. Nat Rev Immunol 20022116126
  136. 136. JackL. JMatherI. HCloning and analysis of cDNA encoding bovine butyrophilin, an apical glycoprotein expressed in mammary tissue and secreted in association with the milk-fat globule membrane during lactation. J Biol Chem 19902651448114486
  137. 137. RybickiB. AWalewskiJ. LMaliarikM. JKianHIannuzziM. CThe BTNL2 gene and sarcoidosis susceptibility in African Americans and whites. Am J Hum Genet 200577491499
  138. 138. SzyldPJagielloPCsernokEGrossW. LEpplenJ. TOn the Wegener granulomatosis associated region on chromosome 621BMC Med Genet 2006
  139. 139. FischerAHofmannSet alA Genome-Wide Linkage Analysis in 181 German Sarcoidosis Families Using Clustered Biallelic Markers, Chest 2010151157
  140. 140. IannuzziM. CIyengarS. KGray-McGuire C, Elston RC, Baughman RP, Donohue JF, Hirst K, Judson MA, Kavuru MS, Maliarik MJ, et al. Genome-wide search for sarcoidosis susceptibility genes in African Americans. Genes Immun 20056509518
  141. 141. Gray-McGuire C., Sinha R, Iyengar SK, Millard C, Rybicki BA, Elston RC, Iannuzzi MC. Genetic characterization and fine mapping of susceptibility genes for sarcoidosis in African Americans on chromosome 5. Hum Genet 2006120420430
  142. 142. ParraE. JMarciniAAkeyJMartinsonJBatzerM. ACooperRForresterTAllisonD. BDekaRFerrellR. Eet alEstimating African American admixture proportions by use of population-specific alleles. Am J Hum Genet 19986318391851
  143. 143. ThompsonC. LRybickiB. AIannuzziM. CElstonR. CIyengarS. KGray-McGuire C. Stratified linkage analysis based on population substructure in a population of African-American sarcoidosis families. Am J Hum Genet 200679603613
  144. 144. HofmannSFrankeAFischerAet alGenome-wide association study identifies ANXA11 as a new susceptibility locus for sarcoidosis. Nat Genet. 200840911031106
  145. 145. HofmannSFischerAet alA genome-wide association study reveals evidence of association with sarcoidosis at 612Eur Respir J 2011
  146. 146. SharpeA. HFreemanG. JTheBCD28 superfamily. Nat Rev Immunol 20022116126
  147. 147. HandaTNagaiSItoITabuenaRShigematsuMHamadaKKitaichiMIzumiTAoyamaTToguchidaJet alPolymorphisms of B7 (CD80 and CD86) genes do not affect disease susceptibility to sarcoidosis. Respiration (Herrlisheim) 200572243248

Written By

Birendra P. Sah and Michael C. Iannuzzi

Submitted: 19 April 2012 Published: 13 March 2013