Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Single Nucleotide Polymorphisms in IL17A and IL6 Are Associated with Decreased Risk for Pulmonary Tuberculosis in Southern Brazilian Population

  • Mariana Milano ,

    marianamilano@hotmail.com (MM); mmoraes@fiocruz.br (MOM)

    Affiliations Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil, Centro de Desenvolvimento Científico e Tecnológico, Fundação Estadual de Produção e Pesquisa em Saúde, Porto Alegre, Rio Grande do Sul, Brazil

  • Milton Ozório Moraes ,

    marianamilano@hotmail.com (MM); mmoraes@fiocruz.br (MOM)

    Affiliation Laboratório de Hanseníase, Instituto Oswaldo Cruz Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil

  • Rodrigo Rodenbusch,

    Affiliation Centro de Desenvolvimento Científico e Tecnológico, Fundação Estadual de Produção e Pesquisa em Saúde, Porto Alegre, Rio Grande do Sul, Brazil

  • Caroline Xavier Carvalho,

    Affiliation Laboratório de Hanseníase, Instituto Oswaldo Cruz Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil

  • Melaine Delcroix,

    Affiliation Division of Infectious Disease and Vaccinology, University of California, Berkeley, United States of America

  • Gabriel Mousquer,

    Affiliation Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil

  • Lucas Laux da Costa,

    Affiliations Centro de Desenvolvimento Científico e Tecnológico, Fundação Estadual de Produção e Pesquisa em Saúde, Porto Alegre, Rio Grande do Sul, Brazil, Programa de Pós-Graduação em Clínica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil

  • Gisela Unis,

    Affiliation Hospital Sanatório Partenon, Porto Alegre, Rio Grande do Sul, Brazil

  • Elis Regina Dalla Costa,

    Affiliation Centro de Desenvolvimento Científico e Tecnológico, Fundação Estadual de Produção e Pesquisa em Saúde, Porto Alegre, Rio Grande do Sul, Brazil

  • Maria Lucia Rosa Rossetti

    Affiliations Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil, Centro de Desenvolvimento Científico e Tecnológico, Fundação Estadual de Produção e Pesquisa em Saúde, Porto Alegre, Rio Grande do Sul, Brazil, Universidade Luterana do Brazil, Canoas, Rio Grande do Sul, Brazil

Abstract

In Mycobacterium tuberculosis (MTB) infection, the complex interaction of host immune system and the mycobacteria is associated with levels of cytokines production that play a major role in determining the outcome of the disease. Several single-nucleotide polymorphisms (SNPs) in cytokine genes have been associated with tuberculosis (TB) outcome. The aim of this study was to evaluate the association between previously reported SNPs IL2–330 T>G (rs2069762); IL4–590 C>T (rs2243250); IL6–174 G>C (rs1800795); IL10–592 A>C (rs1800872); IL10–1082 G>A (rs1800896); IL17A -692 C>T (rs8193036); IL17A -197 G>A (rs2275913); TNF -238 G>A (rs361525); TNF -308 G>A (rs1800629) and IFNG +874 T>A (rs2430561) and pulmonary TB (PTB) susceptibility. We conducted a case-control study in individuals from Southern Brazil who were recruited between February 2012 and October 2013 in a high incidence TB city. We performed a multiplex genotyping assay in 191 patients with PTB and 175 healthy subjects. Our results suggest a decreased risk for PTB development associated with the IL17A -197A allele (OR = 0.29; p = 0.04), AA genotype (OR = 0.12; p = 0.04) and A carrier (AG/AA) (OR = 0.29; p = 0.004) and IL6 -174C carrier (CC/CG) (OR = 0.46; p = 0.04). We could not properly analyze IL17A -692 C>T (rs8193036) and IFNG +874T>A due to genotypic inconsistencies and found no evidence of association for the IL2, IL4, IL10 and TNF polymorphisms and PTB. In conclusion, our results show a protective effect of IL17 and IL6 polymorphisms on PTB outcome in Southern Brazilian population.

Introduction

Tuberculosis (TB) is a chronic granulomatous disease that predominantly affects the lungs and is caused by Mycobacterium tuberculosis (MTB) [1]. TB is ranked as the second leading cause of death from a single infectious agent, and remains a public health issue with 1.5 million of deaths in 2013 [2]. Annually 5.7 million of new TB cases are reported worldwide. Disturbingly, according to World Health Organization approximately one-third of the world’s population is infected with MTB [2].

Consistent with a long lasting host–pathogen interaction and co-evolution, only 10% of MTB-infected individuals progress towards active disease [3]. The mechanisms explaining why some individuals develop the disease are unclear [4]. A significant proportion of interindividual variation in susceptibility to TB can be attributed to environmental factors such as malnutrition, but a substantial part has been credited to host genetic factors [5,6]. Strong evidence derived from family-based, twin and segregation genetic studies indicate that the inherited background influences TB outcome [79]. The critical importance of CD4+ T cell mediated immunity [10,11] and of the interleukin (IL)-12/interferon-gamma pathway in resistance to MTB has been duly described [12,13]. Unexpectedly, genome-wide association studies (GWAS) have shown a limited success finding either genes or pathways associated with TB. These studies were unable to consistently pinpoint major genes and there are no independent replications in the described chromosome regions: 8q, 11p13 and 18q11 [1417]. Nevertheless, genetic determinants of TB susceptibility in case-control studies and in some cases followed by meta-analysis confirmed single nucleotide polymorphisms (SNPs) mostly in immunity-related genes. Studies based on such approaches have properly identified polymorphisms in candidate genes such as IFNG, SLC11A1, IL10, MCP1, and P2RX7 [1823] to be associated with TB susceptibility. For example, IFNG +874T>A the presence of the T allele is associated with TB resistance and higher expression of IFN-gamma [18].

In fact, cytokines play a key role in driving the appropriate immune response to mycobacteria via activation of inflammatory and immunomodulatory networks orchestrated by both macrophages and T cells [24]. The interleukin (IL)-17A is a potent pro-inflammatory cytokine capable of inducing chemokine expression, migration of neutrophil and recruitment and trafficking of Th1 cells to parenchymal tissue during TB infection [24, 25]. IL-17A has also been suggested to be critical in the first steps of TB and granuloma formation [24, 25] along with TNF [26]. Likewise, cytokines as IL-2 and IL-6 are known to work together to promote an immune response satisfactory against MTB [27, 28].

Few studies have investigated the influence of polymorphic variants in IL6 and IL17A genes and PTB outcome showing inconsistent results. The aim of our study was to investigate whether IL6–174 G>C SNP and IL17A -197 G>A polymorphism and additional cytokine genes involved in immune response to MTB (IL2, IL4, IL10, IFNG and TNF) are associated with genetic susceptibility to pulmonary TB (PTB). In this study, the genetic association of polymorphic variants within cytokine genes was evaluated in a case-control study in a PTB population from Southern Brazil.

Materials and Methods

Case–control study

This case-control study recruited 191 PTB patients in a reference hospital for TB in Porto Alegre, the capital of Rio Grande do Sul State in southern Brazil between February 2012 and October 2013.

We enrolled patients > 18 years old, diagnosed with PTB who were on anti-TB and anti-multidrug-resistant (MDR)-TB treatment during any time between disease diagnosis and the completion of treatment. The information about patients’ medical conditions as results of drug susceptibility test, coexisting of forms of extra-pulmonary TB, diabetes, asthma were extracted through medical records. Serum samples were tested for HIV 1/2 and HCV. Patients were confirmed to have PTB by chest X-ray, sputum smear microscopy and culture according recommendations described in Brazilian National TB guideline [29]. Smear sputum samples were tested for MDR-TB at a referral laboratory in Porto Alegre, Brazil [30]. In this study, patients with comorbidities including diabetes, asthma, HCV, HIV or HCV+HIV were included in PTB group and comprised 22% of patients population. Although, some of the comorbidities are known risk factors for TB outcome such as HIV and diabetes, a comparison of patient population stratified according to the presence and absence of comorbidities were not statistically different (data not shown) indicating that comorbidities are unlikely confounding factors. Adjustments in the regression logistic model using comorbidities were not feasible because this condition was present only in patients. We excluded patients who were not in eligible age criteria (≤17 y/o), coexisting of forms of extra pulmonary TB, drug users, prisoners and Native Americans.

The control group was composed by 175 unrelated healthy individuals, over 18 years old, among them 50 household contacts and 125 blood donors recruited at the Hemocentro do Estado do Rio Grande do Sul in Porto Alegre, which is located at the same geographical area that the PTB cases were recruited. All individuals in control group were HIV and HCV or HIV+HCV negatives, without signs or symptoms of TB, previous history of TB or have completed anti-TB prophylaxis. Ethnic background was determined by self-identification as white, non-white, or other. No individual of auto referred Asiatic ancestry participated in the study. All subjects were BCG vaccinated at birth that was confirmed by the presence of the BCG scar.

Ethics considerations

The study was approved by Research Ethics Committee of Fundação Estadual de Produção e Pesquisa em Saúde, FEPPS-RS #09⁄2011 in Porto Alegre, Rio Grande do Sul, Brazil. An information sheet describing the purpose of the study and the individuals’ rights as participants study were read along, and a copy provided, for all volunteers. The researcher also provided a contact phone number for any additional information about the research. After all clarification, a written informed consent was obtained for each person.

SNP genotyping

Genomic DNA was isolated from peripheral blood using Nucleo Spin® Blood kit (Macherey-Nagel Inc.) according to the manufacturer´s instructions. Amplification of regions flanking SNPs in cytokines genes IL2–330 T>G (rs2069762); IL4–590 C>T (rs2243250); IL6–174 G>C (rs1800795); IL10–592 A>C (rs1800872); IL10–1082 G>A (rs1800896); IL17A -197 G>A (rs2275913); IL17A -692 C>T (rs8193036); TNF -238 G>A (rs361525); TNF -308 G>A (rs1800629) and IFNG +874 T>A (rs2430561) was accomplished with a multiplex PCR on 5 to 10 ng genomic DNA, 0.2 μM of each primer (S1 Table) and Qiagen Multiplex PCR Master Mix (QIAGEN,Invitrogen). Amplification reaction consisted of 95°C for 15 minutes followed by 30 cycles of 35 seconds at 94°C, 90 seconds at 57°C, 90 seconds at 72°C and a final extension for 10 minutes at 72°C. Amplicons were purified using Illustra EXOProStar (GE HealthCare®) according to the user’s manual recommendation.

SNP analysis were performed on the SNaPshot® Multiplex System ABI Prism (Applied Biosystems®, São Paulo, Brazil). SNaPshot primers are listed in S1 Table.

According instructions of user's manual the reactions were performed in a final volume of 10 μL, containing 3.0 μL (0.01 to 0.4 ρmol) of purified multiplex PCR product, 5.0 μL of SNaPshot Multiplex Ready Reaction Mix and 1μl of a pooled of the ten SNaPshot primers described at S1 Table. All the ten primers used in the pooled SNaPshot reactions were previously premixed to have a concentration of 1 μM for each primer. Multiplex single base extension was carried out for 28 cycles as follows: 10 seconds at 96°C, 5 seconds at 50°C and 30 seconds at 60°C. SNaPshot products were then incubated at 37°C for 1h with 0.8 U of thermo sensitive alkaline phosphatase (ThermoScientific FastAP). After heat inactivation of the alkaline phosphatase for 20 minutes at 80°C, 1μL of the labeled products were mixed with 9.5 μL of HiDi formamide and 0.5 μL of GeneScan-120 LIZ size standard (Applied Biosystems). They were then separated using an ABIPRISM® 3130xl Genetic Analyzer (Applied Biosystems) with POP-4 polymer and with respective run parameters: injection voltage of 1.2 Kv, injection time of 23 seconds run voltage of 15Kv and run time 1200 seconds in a capillary of 36 cm length. Analysis was performed using Gene Mapper ID software version 3.2.1 (Applied Biosystems). In order to confirm the genotyping system at least 10% of samples were sequenced on ABIPRISM® 3130xl Genetic Analyzer (Applied Biosystems).

Statistical Analysis

All statistical analysis was performed using Genetics package—software R version 2.11.1 [31]. Genotype frequencies were tested for Hardy Weinberg Equilibrium (HWE) using a Chi-square test. Frequencies of the genotypes, alleles and carriers were compared in cases and controls by logistic regression with adjustment for gender, income and age (> or < 25 years old). Income was included as a variable for correction since it was associated with TB outcome and could also be associated with ancestry (genetic variability). Categorical age criteria have been set according epidemiological features of TB [32]. Odds ratio (OR) was used as the point estimates of risk and were calculated along with their 95% confidence intervals (CIs) and the significant p values was adjusted by false discovery rate (FDR) test.

Results

Characteristics of PTB patients and healthy controls

Three-hundred sixty six individuals of the metropolitan area of Porto Alegre, Brazil were included in the study (Table 1). This city it is ranked as the Brazilian capital with the highest incidence of TB [33]. The PTB group was comprised of 191 individuals, 56 females and 135 males, with a mean age of 42.46 (±14.57). Regarding ethnicity within the PTB group, 134 individuals reported themselves as white and 56 as non-whites. However, this covariate showed no association with outcome in logistic regression model, so it was excluded of the analysis. Fifty-six subjects were infected with MDR-TB and 42 had comorbidities such as asthma, diabetes, hepatitis C, HIV/AIDS or HCV+HIV. The healthy control group included 175 individuals, 81 females and 94 males, with a mean age of 36.64 (±14.59) and 133 individuals declared themselves as white and 42 as non-white.

To confirm whether we could run our analysis combining household contacts and blood bank donors as one group defined as healthy controls, we first compared all SNPs in both populations and no difference was observed (S2 Table). Likewise, we compared patients infected with either drug sensitive or resistant strains (S3 Table) and no differences were observed. Thus, all following analyses were run comparing two groups: a combined PTB group and a combined healthy control group.

Cases and controls were in accordance with HWE (data not shown) for the SNPs evaluated except for the IL17A -692 C>T that deviated from HWE in cases and controls and IFNG +874T>A in control group. In order to avoid bias in our analysis IL17A -692 C>T and IFNG +874T>A were excluded of this study.

SNPs in IL17A -197G>A and IL6 -174G>A are associated with protection to PTB development

As observed in Table 2, for—IL17A 197A>G polymorphism the presence of -197A allele was significantly higher in control group than PTB patients (p = 0.04). Furthermore, the logistic regression model showed a decreased risk for PTB development among individuals with AA genotype (adjusted OR = 0.12; p = 0.04) and carriers of A allele (AG+AA) (adjusted OR = 0.29; p = 0.004).

thumbnail
Table 2. Distribution of IL17A -197G>A and IL6 -174G>C Alleles, Genotypes and Carrier Frequencies in PTB Patients and Healthy Controls.

https://doi.org/10.1371/journal.pone.0147814.t002

Regarding IL6 -174G>C polymorphism (Table 2), the CG genotype and carriers of C allele (CC+CG) exhibited a decreased risk of PTB development (OR = 0.55, p = 0.04 and OR = 0.49, p = 0.008 respectively). Heterozygotes for -174 (GC) became not statistically significant after statistical adjustment (OR = 0.54, p = 0.20),

Taken together our results suggest that polymorphisms of IL17A and IL6 cytokine genes at positions -197 and -174, respectively, are associated with protection for PTB development in the Southern Brazilian population.

SNPs in IL2–330 T>G, IL4–590 C>T, IL10–592 A>C, IL10–1082 G>A, TNF -238 G>A and -308 G>A are not associated with to PTB development

Among polymorphisms at positions IL2–330 T>G, IL4–590 C>T, IL10–592 A>C, IL10–1082 G>A, TNF -238 G>A and TNF -308 G>A were found no evidence of association for genotypes, alleles and allele carriers and PTB development (S4 Table). Thus, our results suggest that the SNPs of cytokine genes IL2, IL4, IL10 and TNF above are not directly related to PTB outcome.

Discussion

Genetic effect on TB susceptibility is more complicated than many other common human diseases because there are potential confounding environmental factors involved in TB outcome [34]. However, considerable evidence has accumulated to support the role of human genetic factors in TB susceptibility [79]. In the past few years, population case-control studies have reported association between genetic variations in immune-related genes and susceptibility to TB [18, 20]. SNPs in cytokine genes are able to influence the cytokine levels and regulate resistance and susceptibility to TB [35,36].

The IL-17 cytokine family includes several cytokines among which IL-17A is thought a main pro-inflammatory cytokine being important to the innate and adaptive immune responses [37]. Recent progress in studies of IL-17A cytokine has revealed its important role in protective mechanisms against infectious diseases [24, 25, 38] and in studies of BCG-induced immunity [3940].

The present study shows association between the -197G>A polymorphism and PTB susceptibility in Southern Brazilian population. Our results revealed that -197A allele that was associated with TB, control higher levels of IL-17A release, regulating promoter activity through affinity to transcriptional factor NFAT [41, 42]. Therefore, increased production of IL-17A could be beneficial in to hamper the mycobacterial infection contributing in formation of mature granuloma and blocking disease progression [43, 44].

The IL17A polymorphism (rs2275913) was also evaluated in distinct populations reporting contradictory results. In Northern Spain population, the G allele (OR = 1.40, p = 0.02) and GG genotype (OR = 1.59, p = 0.015) were associated with increased risk for PTB development [45] whereas two other studies failed in replicate such association in Croatian and a Chinese populations [46, 47]. In contrast, a recent report showed A allele carrier (GA/AA) (OR = 1.52, p = 0.006) and AA genotype (OR = 2.2, p = 0.001) associated to increased risk for PTB development [48]. However, this latter study did not fulfill HWE criteria for cases. Therefore, these results should be evaluated with caution. Our findings are converging with the literature suggesting the A allele of IL17A (rs2275913) SNP is associated with TB protection or G allele is associated with TB susceptibility, which means rigorously the same.

Interestingly, the A allele of IL17A (rs2275913) has been associated with increased risk for inflammatory diseases [49,50] indicating that its overproduction could as well be deleterious leading to pathological conditions. However, in MTB infection is likely that a genetic variation slightly increasing IL-17A production may be advantageous in the formation of mature granulomas making difficult mycobacteria spread [44].

The importance of cytokine IL-6 have been shown studies murine models of mycobacterial infections as well in vaccination with a tuberculosis subunit vaccine demonstrating that IL-6 is needed for optimal T-cell development [51, 52]. Indeed, high levels of IL-6 cytokine are produced in response to MTB infection [53, 54] and its role seems especially critical when bacterial burden is high [55]. Reports about the involvement of polymorphic variants within IL6 gene and PTB susceptibility remain inconsistent [56]. To provide further investigation into controversial points between IL6 polymorphisms and TB susceptibility we analyzed the influence of IL6–174 G>C polymorphism on PTB outcome. Our results suggested decreased risk for PTB development among carrier of C allele. The results corroborated previous findings among Pakistani population and confirmed by meta-analysis study [56, 57]. It was previously demonstrated that -174G allele is associated with higher production levels of IL-6 [58]. In this context, it was recognized that during MTB infection higher levels of IL-6 affect the production of other critical cytokines of immune response, such as IFN-γ [59]. Therefore, the decreased frequency of high IL-6 production genotype observed among the controls could be favorable for optimal macrophage activation and effective immunity in mycobacterial infection.

In contrast, the present study showed that IL2–330 T>G is not associated with PTB susceptibility, although the role of IL-2 cytokine in modulating the immune response in TB has become evident. Similarly, it was found no evidence of association between PTB and IL4 -590C>T, TNF -308G>A and TNF -238G>A polymorphisms. Our results corroborate with previous reports concerning TB susceptibility in non-Brazilian populations [18, 60, 61], although, recently the -308A was associated with PTB susceptibility on an ethnically distinct population in Mozambique [62]. Also, the IL4 -590CC genotype showed a protective effect for PTB in a Southern India population [63]. Concerning our results for IL10–1082 and -592, we found no influence of these polymorphisms on PTB outcome. Our result for IL10 polymorphism at -1082 position corroborates with a meta-analysis study in a worldwide-pooled population [18].

Interestingly, more recent meta-analysis studies that performed subgroup analysis for the IL10 -1082G>A SNP revealed conflicting results for analysis stratified by ethnicity for European and American population [20, 64]. Further, according to our findings, IL10–592 A>C analysis showed no risk association with PTB and a meta-analysis study confirmed such finding for Europeans, Africans, and Americans populations but reported a decreased risk for TB development among the Asian population [20]. In this context, reports have highlighted the importance of haplotype analysis since both polymorphisms -592 A>C and -819 C>T in IL10 promoter region are in perfect linkage disequilibrium [64, 65].

The use of popular case-control designs to test candidate genes or genome-wide association studies have proved suitable to pinpoint functional genetic variants in complex diseases [66]. Sometimes it is necessary to test several independent population to replicate the data, but results could be useful to envisage better diagnostics and prognostics methods [66, 67]. TB used to be a chronic disease and it is likely that a subtle balance of immune response could have a great impact over its control and progression. In this regard, our study showed for the first time in that population the protective role of IL17A -197G>A and IL6 -174G>C on PTB outcome. Indeed, our findings support IL17A (rs2275913) and IL6 (rs1800795) polymorphisms as promising markers for PTB risk and indicate the role of IL-6 and IL-17A cytokines in immunophysiology of TB. This novel information of host response to MTB infection can help design more effective vaccines, identify risk populations and new treatment strategies. In the future, additional larger studies are needed to validate our findings. Moreover, a dense SNP mapping on IL6 and IL17A loci could be interestingly for better elucidate such association.

Supporting Information

S1 Table. Primer Sequences for Conventional PCR and for SNaPshot Multiplex System.

https://doi.org/10.1371/journal.pone.0147814.s001

(DOCX)

S2 Table. Allelic and Genotypic Frequencies for Cytokine SNPs in Blood Donors and Household Contacts.

https://doi.org/10.1371/journal.pone.0147814.s002

(DOCX)

S3 Table. Allelic and Genotypic Frequencies for Cytokine SNPs in Pulmonary Tuberculosis Patients Sensitive and Multidrug-Resistant Infected.

https://doi.org/10.1371/journal.pone.0147814.s003

(DOCX)

S4 Table. Allelic, Genotypic and Carrier Frequencies of Cytokine SNPs in Pulmonary Tuberculosis Cases and Healthy Controls.

https://doi.org/10.1371/journal.pone.0147814.s004

(DOCX)

Acknowledgments

We thank all patients, household contacts and blood donors for participating in study and collaborating with this research. We also grateful workers from Hospital Sanatório Partenon and Hemocentro do Estado do Rio Grande do Sul for allowing us interrupt their tough routines over two years of blood collect in their work places.

Author Contributions

Conceived and designed the experiments: MM GU ERDC MLRR GM LLC. Performed the experiments: MM RR GM LLC. Analyzed the data: MOM CXC MM MD. Contributed reagents/materials/analysis tools: MD MM RR ERDC. Wrote the paper: MM MOM.

References

  1. 1. Fernando SL, Britton WJ. Genetic susceptibility to mycobacterial disease in humans. Immunol Cell Biol. 2006;84(2):125–37. pmid:16519730
  2. 2. World Health Organization. Global Tuberculosis Report 2014. Geneva, Switzerland: 2014.
  3. 3. Karlsson EK, Kwiatkowski DP, Sabeti PC. Natural selection and infectious disease in human populations. Nat Rev Genet. 2014;15(6):379–93. pmid:24776769
  4. 4. Barry CE, Boshoff HI, Dartois V, Dick T, Ehrt S, Flynn J, et al. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat Rev Microbiol. 2009;7(12):845–55. pmid:19855401
  5. 5. Berrington WR, Hawn TR. Mycobacterium tuberculosis, macrophages, and the innate immune response: does common variation matter? Immunol Rev. 2007;219:167–86. pmid:17850489
  6. 6. Comstock G. Tuberculosis in twins: a re-analysis of the Prophit Study. Am Rev Resp Dis.1978;117:621–624. pmid:565607
  7. 7. Puffer R. Familial susceptibility to tuberculosis; Its importance as a public health problem. Harvard University Press, Cambridge. 1944;106 pp.
  8. 8. Kallmann FJ, Reisner D. Twin studies on the significance of genetic factors in tuberculosis. Am Rev Tuberc.1942;47:549–574.
  9. 9. Cobat A, Gallant CJ, Simkin L, Black GF, Stanley K, Hughes J, et al. Two loci control tuberculin skin test reactivity in an area hyperendemic for tuberculosis. J Exp Med. 2009;206(12):2583–91. pmid:19901083
  10. 10. Cooper AM. Cell-mediated immune responses in tuberculosis. Annu Rev Immunol. 2009;27:393–422. pmid:19302046
  11. 11. North RJ, Jung YJ. Immunity to tuberculosis. Annu Rev Immunol. 2004;22:599–623. pmid:15032590
  12. 12. Alcaïs A, Fieschi C, Abel L, Casanova JL. Tuberculosis in children and adults: two distinct genetic diseases. J Exp Med. 2005;202(12):1617–21. pmid:16365144
  13. 13. Alcaïs A, Quintana-Murci L, Thaler DS, Schurr E, Abel L, Casanova JL. Life-threatening infectious diseases of childhood: single-gene inborn errors of immunity? Ann N Y Acad Sci. 2010;1214:18–33. pmid:21091717
  14. 14. Baghdadi JE, Orlova M, Alter A, Ranque B, Chentoufi M, Lazrak F, et al. An autosomal dominant major gene confers predisposition to pulmonary tuberculosis in adults. J Exp Med. 2006;203(7):1679–84. pmid:16801399
  15. 15. Thye T, Owusu-Dabo E, Vannberg FO, van Crevel R, Curtis J, Sahiratmadja E, et al. Common variants at 11p13 are associated with susceptibility to tuberculosis. Nat Genet. 2012;44(3):257–9. pmid:22306650
  16. 16. Chimusa ER, Zaitlen N, Daya M, Möller M, van Helden PD, Mulder NJ, et al. Genome-wide association study of ancestry-specific TB risk in the South African Coloured population. Hum Mol Genet. 2014;23(3):796–809. pmid:24057671
  17. 17. Thye T, Vannberg FO, Wong SH, Owusu-Dabo E, Osei I, Gyapong J, et al. Genome-wide association analyses identifies a susceptibility locus for tuberculosis on chromosome 18q11.2. Nat Genet. 2010;42(9):739–41. pmid:20694014
  18. 18. Pacheco AG, Cardoso CC, Moraes MO. IFNG +874T/A, IL10 -1082G/A and TNF -308G/A polymorphisms in association with tuberculosis susceptibility: a meta-analysis study. Hum Genet. 2008;123(5):477–84. pmid:18414898
  19. 19. Li X, Yang Y, Zhou F, Zhang Y, Lu H, Jin Q, et al. SLC11A1 (NRAMP1) polymorphisms and tuberculosis susceptibility: updated systematic review and meta-analysis. PLoS One. 2011;6(1):e15831. pmid:21283567
  20. 20. Liang B, Guo Y, Li Y, Kong H. Association between IL-10 gene polymorphisms and susceptibility of tuberculosis: evidence based on a meta-analysis. PLoS One. 2014;9(2):e88448. pmid:24523896
  21. 21. Flores-Villanueva PO, Ruiz-Morales JA, Song CH, Flores LM, Jo EK, Montaño M, et al. A functional promoter polymorphism in monocyte chemoattractant protein-1 is associated with increased susceptibility to pulmonary tuberculosis. J Exp Med. 2005;202(12):1649–58. pmid:16352737
  22. 22. Tian G, Li X, Li H, Wang X, Cheng B. Systematic meta-analysis of the association between monocyte chemoattractant protein-1 -2518A/G polymorphism and risk of tuberculosis. Genet Mol Res. 2015;14(2):5501–10. pmid:26125747
  23. 23. Wu G, Zhao M, Gu X, Yao Y, Liu H, Song Y. The effect of P2X7 receptor 1513 polymorphism on susceptibility to tuberculosis: A meta-analysis. Infect Genet Evol. 2014;24:82–91. pmid:24650918
  24. 24. Torrado E, Cooper AM. IL-17 and Th17 cells in tuberculosis. Cytokine Growth Factor Rev. 2010;21(6):455–62. pmid:21075039
  25. 25. Umemura M, Yahagi A, Hamada S, Begum MD, Watanabe H, Kawakami K, et al. IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette-Guerin infection. J Immunol. 2007;178(6):3786–96. pmid:17339477
  26. 26. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, Lowenstein CJ, et al. Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity. 1995;2(6):561–72. pmid:7540941
  27. 27. Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol. 2001;19:93–129. pmid:11244032
  28. 28. Khanna M, Srivastava LM, Kumar P. Defective interleukin-2 production and interleukin-2 receptor expression in pulmonary tuberculosis. J Commun Dis. 2003;35(2):65–70. pmid:15562950
  29. 29. Ministério da Saúde. Manual Nacional de recomendações para o controle da tuberculose no Brasil. MS. 2011. Available: http://www.cve.saude.sp.gov.br/htm/TB/mat_tec/manuais/MS11_Manual_Recom.pdf
  30. 30. World Health Organization. Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis: 2011 Update. Geneva, Switzerland: WHO.
  31. 31. Team RDC. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing, 2008.
  32. 32. Alcaïs A, Fieschi C, Abel L, Casanova JL. Tuberculosis in children and adults: two distinct genetic diseases. J Exp Med. 2005;202(12):1617–21. pmid:16365144
  33. 33. Ministério da Saúde. Boletim Epidemiológico. Secretaria da Saúde.-MS. Available: http://portalsaude.saude.gov.br/images/pdf/2014/maio/06/boletim2-2013-tb-web.pdf
  34. 34. Stein CM, Baker AR. Tuberculosis as a complex trait: impact of genetic epidemiological study design. Mamm Genome. 2011;22(1–2):91–9. pmid:21104256
  35. 35. Fairfax BP, Knight JC. Genetics of gene expression in immunity to infection. Curr Opin Immunol. 2014;30:63–71. pmid:25078545
  36. 36. O'Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, Berry MP. The immune response in tuberculosis. Annu Rev Immunol. 2013;31:475–527. pmid:23516984
  37. 37. Isailovic N, Daigo K, Mantovani A, Selmi C. Interleukin-17 and innate immunity in infections and chronic inflammation. J Autoimmun. 2015;60:1–11. pmid:25998834
  38. 38. Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immunity. 2011;34(2):149–62. pmid:21349428
  39. 39. Wareham AS, Tree JA, Marsh PD, Butcher PD, Dennis M, Sharpe SA. Evidence for a role for interleukin-17, Th17 cells and iron homeostasis in protective immunity against tuberculosis in cynomolgus macaques. PLoS One. 2014; 9 (2):e88149. pmid:24505407
  40. 40. Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, Cilley GE, et al. IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat Immunol. 2007;8(4):369–77. pmid:17351619
  41. 41. Liu XK, Lin X, Gaffen SL. Crucial role for nuclear factor of activated T cells in T cell receptor-mediated regulation of human interleukin-17. J Biol Chem. 2004;279(50):52762–71. pmid:15459204
  42. 42. Espinoza JL, Takami A, Nakata K, Onizuka M, Kawase T, Akiyama H, et al. A genetic variant in the IL-17 promoter is functionally associated with acute graft-versus-host disease after unrelated bone marrow transplantation. PLoS One. 2011;6(10):e26229. pmid:22028838
  43. 43. Gopal R, Monin L, Slight S, Uche U, Blanchard E, Fallert Junecko BA, et al. Unexpected role for IL-17 in protective immunity against hypervirulent Mycobacterium tuberculosis HN878 infection. PLoS Pathog. 2014;10(5):e1004099. pmid:24831696
  44. 44. Etna MP, Giacomini E, Severa M, Coccia EM. Pro- and anti-inflammatory cytokines in tuberculosis: a two-edged sword in TB pathogenesis. Semin Immunol. 2014;26(6):543–51. pmid:25453229
  45. 45. Ocejo-Vinyals JG, de Mateo EP, Hoz M, Arroyo JL, Agüero R, Ausín F, et al. The IL-17 G-152A single nucleotide polymorphism is associated with pulmonary tuberculosis in northern Spain. Cytokine. 2013;64(1):58–61. pmid:23778030
  46. 46. Bulat-Kardum LJ, Etokebe GE, Lederer P, Balen S, Dembic Z. Genetic Polymorphisms in the Toll-like Receptor 10, Interleukin (IL)17A and IL17F Genes Differently Affect the Risk for Tuberculosis in Croatian Population. Scand J Immunol. 2015;82(1):63–9. pmid:25857634
  47. 47. Peng R, Yue J, Han M, Zhao Y, Liu L, Liang L. The IL-17F sequence variant is associated with susceptibility to tuberculosis. Gene. 2013;515(1):229–32. pmid:23219503
  48. 48. Shi GC, Zhang LG. Influence of interleukin-17 gene polymorphisms on the development of pulmonary tuberculosis. Genet Mol Res. 2015;14(3):8526–31. pmid:26345782
  49. 49. Arisawa T, Tahara T, Shibata T, Nagasaka M, Nakamura M, Kamiya Y, et al. The influence of polymorphisms of interleukin-17A and interleukin-17F genes on the susceptibility to ulcerative colitis. J Clin Immunol. 2008;28(1):44–9. pmid:17828618
  50. 50. Chen J, Deng Y, Zhao J, Luo Z, Peng W, Yang J, et al. The polymorphism of IL-17 G-152A was associated with childhood asthma and bacterial colonization of the hypopharynx in bronchiolitis. J Clin Immunol. 2010;30(4):539–45. pmid:20437253
  51. 51. Appelberg R, Castro AG, Pedrosa J, Minóprio P. Role of interleukin-6 in the induction of protective T cells during mycobacterial infections in mice. Immunology. 1994;82(3):361–4. pmid:7959868
  52. 52. Leal IS, Smedegârd B, Andersen P, Appelberg R. Interleukin-6 and interleukin-12 participate in induction of a type 1 protective T-cell response during vaccination with a tuberculosis subunit vaccine. Infect Immun. 1999;67(11):5747–54. pmid:10531224
  53. 53. Giacomini E, Iona E, Ferroni L, Miettinen M, Fattorini L, Orefici G, et al. Infection of human macrophages and dendritic cells with Mycobacterium tuberculosis induces a differential cytokine gene expression that modulates T cell response. J Immunol. 2001;166(12):7033–41. pmid:11390447
  54. 54. Indrigo J, Hunter RL, Actor JK. Influence of trehalose 6,6'-dimycolate (TDM) during mycobacterial infection of bone marrow macrophages. Microbiology. 2002;148(Pt 7):1991–8. pmid:12101287
  55. 55. Ladel CH, Blum C, Dreher A, Reifenberg K, Kopf M, Kaufmann SH. Lethal tuberculosis in interleukin-6-deficient mutant mice. Infect Immun. 1997;65(11):4843–9. pmid:9353074
  56. 56. Mao X, Ke Z, Liu S, Tang B, Wang J, Huang H, et al. IL-1β +3953C/T, -511T/C and IL-6 -174C/G polymorphisms in association with tuberculosis susceptibility: A meta-analysis. Gene. 2015.
  57. 57. Ansari A, Hasan Z, Dawood G, Hussain R. Differential combination of cytokine and interferon- γ +874 T/A polymorphisms determines disease severity in pulmonary tuberculosis. PLoS One. 2011;6(11):e27848. pmid:22140472
  58. 58. Fishman D, Faulds G, Jeffery R, Mohamed-Ali V, Yudkin JS, Humphries S, et al. The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J Clin Invest. 1998;102(7):1369–76. pmid:9769329
  59. 59. Nagabhushanam V, Solache A, Ting LM, Escaron CJ, Zhang JY, Ernst JD. Innate inhibition of adaptive immunity: Mycobacterium tuberculosis-induced IL-6 inhibits macrophage responses to IFN-gamma. J Immunol. 2003;171(9):4750–7. pmid:14568951
  60. 60. Möller M, Nebel A, van Helden PD, Schreiber S, Hoal EG. Analysis of eight genes modulating interferon gamma and human genetic susceptibility to tuberculosis: a case-control association study. BMC Infect Dis. 2010;10:154. pmid:20525402
  61. 61. Zhang Z, Zhu H, Pu X, Meng S, Zhang F, Xun L, et al. Association between tumor necrosis factor alpha-238G/a polymorphism and tuberculosis susceptibility: a meta-analysis study. BMC Infect Dis. 2012;12:328. pmid:23192010
  62. 62. Mabunda N, Alvarado-Arnez LE, Vubil A, Mariamo A, Pacheco AG, Jani IV, et al. Gene polymorphisms in patients with pulmonary tuberculosis from Mozambique. Mol Biol Rep. 2015;42(1):71–6. pmid:25239251
  63. 63. Vidyarani M, Selvaraj P, Prabhu Anand S, Jawahar MS, Adhilakshmi AR, Narayanan PR. Interferon gamma (IFNgamma) & interleukin-4 (IL-4) gene variants & cytokine levels in pulmonary tuberculosis. Indian J Med Res. 2006;124(4):403–10. pmid:17159260
  64. 64. Gao X, Chen J, Tong Z, Yang G, Yao Y, Xu F, et al. Interleukin-10 promoter gene polymorphisms and susceptibility to tuberculosis: a meta-analysis. PLoS One. 2015;10(6):e0127496. pmid:26030829
  65. 65. Alvarado-Arnez LE, Amaral EP, Sales-Marques C, Durães M B S, Cardoso CC, Nunes Sarno E, et al. Association of IL10 Polymorphisms and Leprosy: A Meta-Analysis. PLoS One. 2015;10(9):e0136282. pmid:26340474
  66. 66. Moraes MO, Pacheco AG.Genetics of complex diseases: knowing gene polymorphisms do matter. Cad Saude Publica. 2013; 29(11):2144–6. pmid:24233030
  67. 67. Cardoso CC, Pereira AC, de Sales Marques C, Moraes MO. Leprosy susceptibility: genetic variations regulate innate and adaptive immunity, and disease outcome. Future Microbiol. 2011 May;6(5):533–49. pmid:21585261