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

A Functional Promoter Polymorphism of IFITM3 Is Associated with Susceptibility to Pediatric Tuberculosis in Han Chinese Population

  • Chen Shen ,

    Contributed equally to this work with: Chen Shen, Xi-rong Wu

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Xi-rong Wu ,

    Contributed equally to this work with: Chen Shen, Xi-rong Wu

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Wei-wei Jiao,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Lin Sun,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Wei-xing Feng,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Jing Xiao,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Qing Miao,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Fang Liu,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Qing-qin Yin,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • Chen-guang Zhang,

    Affiliation Department of Cell Biology, Capital Medical University, Beijing, China

  • Ya-jie Guo,

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

  • A-dong Shen

    shenad16@hotmail.com

    Affiliation Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China

Abstract

A susceptibility locus for tuberculosis, a re-emerging infectious disease throughout the world, was previously discovered to exist on chromosome 11p15. IFITM3 gene encoding for interferon inducible transmembrane protein 3, is located at 11p15. It acts as an effector molecule for interferon-gamma, which is essential for anti-tuberculosis immune response. In order to investigate the association between susceptibility to TB and genetic polymorphisms of the IFITM3 core promoter, a case-control study including 368 TB patients and 794 healthy controls was performed in Han Chinese children in northern China. The rs3888188 polymorphism showed significant association with susceptibility to TB. The rs3888188 G allele, acting recessively, was more frequent in TB patients (95% confidence interval: 1.08–1.56, Bonferroni P-value: 0.039). We further assessed the effect of rs3888188 polymorphism on IFITM3 transcription in vitro. As based on luciferase promoter assays, the promoter activity of haplotypes with rs3888188 G allele was lower than that of haplotypes with rs3888188 T allele. Moreover, peripheral-blood mononuclear cells carrying rs3888188 GG genotype showed a reduced IFITM3 mRNA level compared to cells carrying TT or GT genotype. In conclusion, rs3888188, a functional promoter polymorphism of IFITM3, was identified to influence the risk for pediatric TB in Han Chinese population.

Introduction

Tuberculosis (TB),with Mycobacterium tuberculosis (M. tuberculosis) as the major pathogen, is still a major threat to worldwide human health. M. tuberculosis infects approximately one-third of the world's population. However, only about 10% of infected individuals eventually develop TB disease, whilst the majority of humans are naturally resistant to TB, indicating that host genetic factors play an essential role in determining TB susceptibility [1], [2]. Genetic variations in an increasing number of genes have been associated with human susceptibility to TB disease [3][6].

In China, the prevalence of TB in adults is 1.04‰ by WHO report 2012 [7], while the prevalence of TB in children is 0.918‰ according to a nationwide survey [8]. Compared with adults, children present a special risk group for TB due to rapid progression of a recent infection towards disease [9][12]. Certain pediatric TB reflects Mendelian predispositions, while adult TB seems to be more complex for genetic predisposition [11]. Studying the effects of the candidate susceptibility genes on pediatric TB may aid in the establishment of more efficient prevention of TB spread.

Interferon inducible transmembrane protein 3 (IFITM3) is a double trans-membrane protein that can be upregulated by interferons. High expression of IFITM3 has been detected in periphery leukocytes, HeLa cell lines, and other cells and tissues [13][20]. IFITM3 participates in interferon-triggered processes, such as homotypic cell adhesion, anti-proliferative activities in tumor pathogenesis, and the innate immune response to virus infections [13][19]. Interferon gamma (IFN-γ) is produced and released in response to the presence of pathogens [21], it is secreted as a major cytokine to activate macrophages during M. tuberculosis infection [22]. Mutant mice lacking Ifng have weakened defense against M. tuberculosis [23], and genetic variations in IFNG and its receptor, IFNGR, have been implicated in susceptibility to human TB [24], [25]. IFITM3 has been proven to be an important component of IFN-γ signaling pathway, as down-regulation of IFITM3 via siRNA could largely reduce the anti-virus activities conducted by IFN-γ [18]. Thus, IFITM3 is a potential candidate gene for TB susceptibility.

The IFITM3 gene is located on chromosome 11p15. In this regard, it is worth mentioning that Stein et al. previously found in a genome wide scan that one of the TB-linked loci was located in this chromosome region [26]. The core promoter region of human IFITM3 is enriched with regulatory elements (such as ISRE elements, the SP1-binding motif, and other regulatory sequences), and these are thought to be critical for its transcription [27][30]. In this current study, we investigated the relationship between IFITM3 and susceptibility to TB via polymorphism analysis of its core promoter region around transcription start site. To determine whether or not these IFITM3 polymorphisms are associated with susceptibility to TB, its alleles and genotypes were analyzed in case-control study in Han Chinese pediatric population.

Materials and Methods

Ethics statement

Clinical investigation had been conducted according to the principles expressed in the Declaration of Helsinki. This research has been approved by the Ethics Committee of Beijing Children's Hospital. Written informed consent was obtained from the patients or the guardians of the patients that participated in this research.

Study sample

All the participants involved in this research were Han ethnicity and originated from Northern China (Beijing city and surrounding provinces). They were vaccinated with BCG at birth, which was confirmed by the presence of a scar on the shoulder and by their vaccination records. The pediatric TB patients (n = 368) were newly diagnosed to be Pulmonary TB (PTB, pathological changes limited to in lung) or Extra-pulmonary TB (EPTB, pathological changes involving other tissues) by at least two experienced pediatricians in Beijing Children's Hospital according to the pediatric TB clinical diagnosis standard [31][33]. The gold standard diagnosis of pediatric TB is made when M. tuberculosis is isolated. There were 42 bacteriology confirmed TB patients, in addition to their clinical signs/symptoms suggestive of TB, at least 1 M. tuberculosis positive culture from their sputum, which could be sampled from expectorated sputum, induced sputum, nasopharyngeal aspirates, gastric aspirates, or string tests (or other relevant intrathoracic samples) were obtained. In the absence of bacteriological confirmation, the diagnosis of pediatric TB is based on a triad of (1) clinical presentation (symptoms or signs); (2) imaging (chest radiography and computed tomography scan); (3) contact history (family, close contact); (4) purified protein derivative (PPD) tuberculin skin test (using 5 TU); (5) positive clinical response to anti-TB therapy;(6)Except other lung disease, such as the various reasons pneumonia, lung tumor and so on. Clinical TB could be diagnosed if positive features of (1) plus (2) or (1) plus either two of (3)–(6) were present.

Participants of the control group (n = 794) were recruited among those admitted to Beijing Children's Hospital for surgery. All of them had negative tuberculin PPD skin-test results (<5 mm) and no history of TB or HIV infection, and were matched with TB cases for age, sex, and ethnicity.

DNA extraction and genotyping.

Genomic DNA was extracted from peripheral leukocytes by using a Genomic DNA Extraction kit (QIAamp DNA Blood Mini Kit; Qiagen, Hilden, Germany). The core promoter of IFITM3 was amplified by PCR using 5′-GAG CCC TGA ACC GGG ACA GTG-3′ and 5′-TGG TGT CCA GCG AAG ACC AGC-3′ primers and genotyped by sequencing using a 3730 DNA Analyzer (Applied Biosystems, Foster City, CA, USA).

Plasmid constructs.

Forward (5′-TAT ACT GCA GCT AGC GAG CCC TGA ACC GGG ACA GTG-3′) and reverse (5′-TAT ACT GCA CTC GAG TGG TGT CCA GCG AAG ACC AGC-3′) primers were used to amplify the three most common promoter haplotypes of IFITM3. Promoter haplotypes were independently inserted between the NheI and XhoI restriction sites of the pGL3-based plasmid (Promega, Madison, WI, USA) to generate the pGL3-IFITM3 plasmids. Site-directed mutagenesis of the -204 G/T (rs3888188) site of the pGL3-IFITM3 plasmids was carried out using a QuickChange site-directed mutagenesis kit (Stratagene, La Jolla, CA, USA) following the manufacturer's protocol. This process generated three new constructs with the −204 G/T site changed to −204 T/G. These mutations were confirmed by DNA sequencing.

Cell culture and luciferase assays

HeLa cells were maintained in Dulbecco's modified Eagle's medium (Gibco BRL, USA) supplemented with 10% fetal bovine serum, penicillin (100 U/ml), and streptomycin (100 U/ml). Each constructed pGL3-IFITM3 plasmid was transfected into HeLa cells with the pRL-SV40 plasmid (Promega, USA) by using Lipofectamine 2000 (Invitrogen, USA). Firefly luciferase and renilla luciferase (both via pRL-SV40 plasmid) activities were sequentially measured by a luminometer 48 h after transfection, with or without IFN-γ treatment (final concentration of 100 pg/ml and 0 h, respectively) over a serial time span (5 h and 20 h), utilizing a Dual-Luciferase reporter assay system (Promega, USA). Results were expressed as relative light units of firefly luciferase activity over relative light units of renilla luciferase activity. All experiments were performed in triplicate and repeated three times.

Expression levels of IFITM3 in individuals with different genotypes

A ficoll gradient density centrifugation method [14] was used for peripheral blood mononuclear cell separation. Cells were resuspended in RPMI medium with 10% fetal bovine serum. Cells (106 in 0.5 ml) were seeded into each well of a 48-well plate and incubated in 5% CO2 at 37°C for 24 h with or without adding IFN-γ (final concentration of 100 pg/ml) over a serial time span (2 h, 8 h, and 24 h). Total RNA was extracted and used for cDNA synthesis. Real-time PCR was performed in an ABI7300 Sequence Detection System (Applied Biosystems, USA) using SYBRPremix Ex Taq II (TaKaRa Bio, Japan). The primers for IFITM3 were as follows: forward: 5′-ATG AAT CAC ACT GTC CAA ACC TTC T-3′ and reverse: 5′-CTA TCC ATA GGC CTG GAA GAT CAG-3′. Primers for the control (18 S) reaction were: forward 5′-GGA AGG GCA CCA CCG GAG T-3′ and reverse 5′-TGC AGC CCC GGA CAT CAA G-3′.

Genetic and Statistical anaylysis

Hardy-Weinberg equilibrium (HWE), Linkage disequilibrium (LD), allele frequency, genotype distribution, and haplotype analyses were performed using the SHEsis program (http://analysis.bio-x.cn/myAnalysis.php) [34]. Haplotype frequencies were inferred using the derived expectation maximization (EM) algorithm as implemented in the SHEsis software. Power and sample size calculations were performed using Quanto 1.2.4 software (http://hydra.usc.edu/gxe)[35]. Statistical analysis was carried out using the Statistical Package for Social Sciences version 13.0 (SPSS, Chicago, IL). Differences between non-contiguous variables, genotype distribution and allele frequency were tested by Fisher's exact test. Odds Ratios (OR) and 95% confidence intervals (CI) were examined by logistic regression analysis. The student's t-test was used to compare the promoter activity between haplotypes or IFITM3 mRNA level between genotypes. All statistical hypothesis tests were two-sided with P-value <0.05 representing statistical significance. Bonferroni corrections were used for multiple tests.

Results

Patients and controls

The mean age was 5.8 y (SD 4.7; range, 2 months–17 y) for TB patients and 6.3 y (SD 4.0; range, 3 months–17 y) for the non-TB control subjects. The proportion of male subjects was 233/368 (63.3%) in the TB patient group, and 520/794(65.5%) in the control group. Pulmonary TB (PTB) and Extra-pulmonary TB (EPTB) were diagnosed in 151 (41.0%) cases and 217 (59.0%) cases respectively. Details are shown in Table 1.

Genotyping and genetic analysis

Eight single-nucleotide polymorphisms (SNPs) were genotyped in the 356 base-pair PCR products of the IFITM3 core promoter region around transcription start site: −291 C>T (rs61876247, 291 base pairs upstream of the translation start site of IFITM3), −242 C>A (rs35409983), −231 C>T (rs28602580), −204 T>G (rs3888188), −188 T>C (rs6598045), −181 C>T (rs7478728), −178 C>A (rs71452596), −175 C>T (rs7479267). All these SNPs were in Hardy-Weinberg equilibrium (P>0.05) both in the TB and control groups. The genotyping results of detected SNPs are summarized in Table 2. Three SNPs had a minor allele frequency (MAF) lower than 5% in both control and TB groups: rs61876247 (3.4% and 2.6%, in TB and control groups, respectively), rs35409983 (1.4% and 0.6% respectively), and rs28602580 (1.8% and 0.6% respectively). Linkage disequilibrium coefficients (|D′|) between all SNP pairs were calculated, and the absolute LD (|D′| = 1 and r2 = 1) was found among three SNPs, rs7478728, rs71452596 and rs7479267.

thumbnail
Table 2. The genotyping results of SNPs in IFITM3 core promoter.

https://doi.org/10.1371/journal.pone.0067816.t002

Genetic association of a SNP rs3888188 with TB disease was observed after an application of the Bonferroni correction for multiple testing. The frequency of G allele in TB group was significantly higher than that in control group (OR: 1.30, 95% CI: 1.08–1.56, Bonferroni P = 0.039). Analysis of genotypic distribution of rs3888188 using 3×2 χ2 test revealed significant difference between TB group and control group (Bonferroni P = 0.013). No statistical differences were observed between PTB and EPTB subgroups (data not shown). Further, a 2×2 χ2 test was used by combining different genotype combinations to test dominant (GG+GT versus TT) and recessive (GG versus GT+TT) models of inheritance. The OR for the G allele of rs3888188 as a possible risk factor was 1.58 (95%CI: 1.32–1.89, P = 5.30e–007) under a recessive model and 1.12 (95% CI: 0.87–1.45, P = 0.384) under a dominant model. Thus, a Mendelian recessive trait was accepted for the inheritance pattern.

As three SNPs (rs61876247, rs35409983, and rs28602580) had an MAF lower than 5% in both control and TB groups and rs7478728 alone could represent rs71452596 and rs7479267 (in absolute LD), Haplotype analysis was performed for rs3888188, rs6598045 and rs7478728. Accepting the lowest frequency threshold for this haplotype analysis to be 0.05, three haplotypes including GTT, TCT, and TTC were identified (Table 3). The frequency of GTT haplotype (carrying rs3888188 G allele) was significantly higher in TB patients than that in controls (OR: 1.31, 95%CI: 1.09–1.58, P = 0.004), while the frequency of TCT haplotype (carrying rs3888188 T allele) was significantly lower in the TB group than that in controls (OR: 0.71, 95% CI: 0.57–0.89, P = 0.003).

thumbnail
Table 3. Haplotypes carried out using rs3888188, rs6598045, and rs7478728 by SHEsis.

https://doi.org/10.1371/journal.pone.0067816.t003

Promoter activities

Three common promoter haplotypes, GTT, TTC, and TCT were amplified from genomic DNA of known homozygous individuals and subcloned into pGL3-basic plasmid construct to generate pGL3-IFITM3 plasmids. Site-directed mutagenesis was then carried out on rs3888188 SNP in each plasmid. Following transfection and subsequent dual luciferase assays, the transcriptional activity of the GTT (carrying rs3888188 G allele) haplotype promoter was significantly lower than that of TTC and TCT (both carrying rs3888188 G allele) promoter (Fig. 1A). Furthermore, the transcriptional activity of the promoter construct with the TB-susceptible rs3888188 G allele was lower than that of the rs3888188 T allele for each pair of haplotypes (before and after site-directed mutagenesis) (Fig. 1B).

thumbnail
Figure 1. Promoter activity and gene expression analysis of IFITM3.

Data are shown as the mean of triplicate determinations ± standard deviation (SD) and are representative of three independent experiments. Significant difference was defined as a P-value <0.05 by two-tailed unpaired t test. (A) Dual luciferase assays for haplotypes composed of rs3888188, rs6598045, and rs7478728: comparison of the relative luciferase activities of the ‘GTT,’ ‘TTC,’ and ‘TCT’ haplotypes; † indicates significant difference in comparisons of both GTT vs. TTC and CTT vs. TCT. (B) Dual luciferase assays for haplotypes before and after site-directed mutagenesis: § indicates a significant difference for GTT vs. TTT, ¶ for GTC vs. TTC, and # for GCT vs. TCT.

https://doi.org/10.1371/journal.pone.0067816.g001

Expression levels of IFITM3 in lymphocytes with different genotype of rs3888188

As a Mendelian recessive trait was accepted for rs3888188 G allele, we further investigated whether rs3888188 GG genotype would influence IFITM3 mRNA expression level. Fifteen healthy individuals (4 with rs3888188 TT genotype, 6 with TG genotype, and 5 with GG genotypes, age 10 ± 3 years) were recruited for this experiment. IFITM3 mRNA analysis was performed in lymphocytes from these 15 individuals. The relative mRNA levels of IFITM3 in all genotypes continued to rise within 2–24 hours after IFN-γ was added. Lymphocytes carrying the rs3888188 GG genotype had significantly lower IFITM3 expression level compared to the cells carrying other genotypes (TT plus TG) (Fig. 2).

thumbnail
Figure 2. Gene expression analysis of IFITM3.

The relative mRNA expression levels of IFITM3 were displayed according to genotypes of rs3888188: Data are shown as the mean determinations ± standard deviation (SD) and are representative of three independent experiments. Significant difference was defined as a P-value <0.05 by two-tailed unpaired t test. * indicates a significant difference as compared to GG genotype.

https://doi.org/10.1371/journal.pone.0067816.g002

Discussion

Children not only have an increased risk of developing progressive disease following exposure, but have a much greater risk of developing disseminated forms of the disease such as tuberculosis meningitis (TBM). Although this could be largely resulted from the immaturity of the immune response, certain pediatric TB reflects Mendelian predispositions, while adult TB seems to be more complex for genetic predisposition [11].

IFITM3 is an important component of the IFN-γ signaling pathway. This has been demonstrated through down-regulation of IFITM3 via siRNA, which reduced IFN-γ antiviral activity by 40–70% [17]. IFN-γ is thought to be essential factor acting against different infectious agents including tuberculosis. Successful immune reaction against M. tuberculosis requires strong IFN-γ signaling. As such, IFITM3 is a potential candidate gene for TB susceptibility. Thus, we speculated that IFITM3 might have a clear influence on the occurrence of TB. Until now, no data on the IFITM3 genetic polymorphisms were published for Han Chinese population. As promoter variation might influence gene expression, we initially investigated the association of polymorphisms in the core promoter region of IFITM3 with TB in a subset of the pediatric population of Han Chinese ethnicity.

This case-control study showed the rs3888188G allele to be linked to the increased risk of TB. In genetic studies, the power of a sample set to detect an association greatly depends on the allele frequency and the size of its effect [34]. In our study, a power to detect an effect for rs3888188 was calculated to be high (99.6%) under the sample size used. The frequency of the rs3888188 G allele in TB group was significantly higher than that in the control group after Bonferroni correction and a recessive model of inheritance was found significant. Besides, the frequency of GTT haplotype (that contains rs3888188 G allele) was significantly higher in TB patients than in controls while the frequency of TTC haplotype (rs3888188 T allele) was significantly lower in TB patients than in controls. Since the IFITM3 gene is located at 11p15.5, these findings may also support the identification of the TB-susceptibility locus at 11p15 suggested by the genome scan [26].

Because IFITM3 plays a pivotal role in IFN-γ signaling and successful immunity against M. tuberculosis requires strong IFN-γ signaling, we further hypothesized that the TB-susceptible rs3888188 G allele could lead to reduced IFITM3 transcription. By dual-luciferase reporter analysis, we found the transcriptional activities of the rs3888188 G allele promoter to be lower than that of the T allele promoter. We also noticed that lymphocytes carrying the rs3888188 GG genotype had significantly lower IFITM3 mRNA level compared with cells carrying other two genotypes (TT plus TG). These observations indicated that insufficient transcriptional activity of IFITM3 could be a risk factor for TB susceptibility and that IFITM3 has a protective role against TB. However, further studies are needed to determine the specific transcription factors involved in the rs3888188-regulated promoter activity.

One possible mechanism by which IFITM3 expression levels may modulate susceptibility to TB is a reduced capacity to eliminate M. tuberculosis in phagolysosome. IFITM3 participates in the lysosomal pathway and was shown to limit viral replication at a late stage of endocytosis [18], as IFITM3 overexpression lead to the expansion of enlarged acidified compartments consisting of lysosomes. Previously, lysosome related genes, such as SLC11A1 (encoding solute carrier family 11 member 1, a lysosome membrane protein) [36] and IRGM (encoding immunity-related GTPase family M, mediating fusion of M. tuberculosis-contained endosome with lysosome) [37], were proven to be associated with TB susceptibility. Lysosomal pathway plays an important role in TB pathogenesis [36], [37], it may directly determine the successful elimination of M. tuberculosis by macrophages or the survival of M. tuberculosis within macrophages. As sufficient expression of IFITM3 effectively restricted infection by influenza virus [16], [17], vesticular stomatitis virus [18], filoviruses, and SARS coronavirus [16], sufficient expression of IFITM3 might also be critical in reducing survival of M. tuberculosis.

The present study was conducted among newly diagnosed pediatric TB patients of Han Chinese origin and healthy controls with the same ethnic background. Association linking IFITM3 to pediatric TB is reported here for the first time and has not been previously investigated in any population.

Acknowledgments

We thank all subjects for their participation in this study.

Author Contributions

Conceived and designed the experiments: CS AS. Performed the experiments: CS XW FL WJ LS WF QY. Analyzed the data: CS XW. Contributed reagents/materials/analysis tools: CS XW JX QM CZ YG. Wrote the paper: CS XW AS.

References

  1. 1. Dye C, Williams BG (2010) The population dynamics and control of tuberculosis. Science 328: 856–856.
  2. 2. Schurr E (2007) Is susceptibility to tuberculosis acquired or inherited? J Intern Med 261(2): 106–11.
  3. 3. Bellamy R (2003) Susceptibility to mycobacterial infections: the importance of host genetics. Genes Immun 4: 4–11.
  4. 4. Bellamy R (2005) Genetic susceptibility to tuberculosis. Clin Chest Med 26: 233–246.
  5. 5. Möller M, Hoal EG (2010) Current findings, challenges and novel approaches in human genetic susceptibility to tuberculosis. Tuberculosis (Edinb) 90: 71–83.
  6. 6. Yim JJ, Selvaraj P (2010) Genetic susceptibility in tuberculosis. Respirology 15: 241–56.
  7. 7. Global tuberculosis report 2012. World Health Organization website. Available: http://www.who.int/tb/publications/global_report/en/index.html. Accessed 2013 May 30.
  8. 8. Jiang ZF, Yi ZW, Zhao SY (2006) Practical Pediatric Tuberculosis. Beijing: People's Medical Publishing House. pp. 431.
  9. 9. Newton SM, Brent AJ, Anderson S, Whittaker E, Kampmann B (2008) Paediatric tuberculosis. Lancet Infect Dis 8: 498–510.
  10. 10. Rigouts L (2009) Clinical practice: diagnosis of childhood tuberculosis. Eur J Pediatr 168: 1285–1290.
  11. 11. Alcais A, Fieschi C, Abel L, Casanova JL (2005) Tuberculosis in children and adults: two distinct genetic diseases. J Exp Med 202: 1617–1621.
  12. 12. Walls T, Shingadia D (2004) Global epidemiology of paediatric tuberculosis. J Infect 48: 13–22.
  13. 13. Seyfried NT, Huysentruyt LC, Atwood JA 3rd, Xia Q, Seyfried TN, et al. (2008) Up-regulation of NG2 proteoglycan and interferon-induced transmembrane proteins 1 and 3 in mouse astrocytoma: a membrane proteomics approach. Cancer Lett 263: 243–252.
  14. 14. Fan J, Peng Z, Zhou C, Qiu G, Tang H, et al. (2008) Gene-expression profiling in Chinese patients with colon cancer by coupling experimental and bioinformatic genomewide gene-expression analyses: identification and validation of IFITM3 as a biomarker of early colon carcinogenesis. Cancer 113: 266–275.
  15. 15. Hisamatsu T, Watanabe M, Ogata H, Ezaki T, Hozawa S, et al. (1999) Interferon-inducible gene family 1-8U expression in colitis-associated colon cancer and severely inflamed mucosa in ulcerative colitis. Cancer Res 59: 5927–5931.
  16. 16. Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, et al. (2009) The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 139: 1243–1254.
  17. 17. Jiang D, Weidner JM, Qing M, Pan XB, Guo H, et al. (2010) Identification of five interferon-induced cellular proteins that inhibit west nile virus and dengue virus infections. . J Virol 84: 8332–8341.
  18. 18. Weidner JM, Jiang D, Pan XB, Chang J, Block TM, et al. (2010) Interferon-induced cell membrane proteins, IFITM3 and tetherin, inhibit vesicular stomatitis virus infection via distinct mechanisms. J Virol 84: 12646–12657.
  19. 19. Feeley EM, Sims JS, John SP, Chin CR, Pertel T, et al. (2011) IFITM3 inhibits influenza A virus infection by preventing cytosolic entry. PLoS Pathog 7: e1002337.
  20. 20. Seo GS, Lee JK, Yu JI, Yun KJ, Chae SC, et al. (2010) Identification of the polymorphisms in IFITM3 gene and their association in a Korean population with ulcerative colitis. Exp Mol Med 42: 99–104.
  21. 21. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD (1998) How cells respond to interferons. Annu Rev Biochem 67: 227–264.
  22. 22. Lee J, Kornfeld H (2010) Interferon-γ Regulates the Death of M. tuberculosis-Infected Macrophages. J Cell Death 3: 1–11.
  23. 23. Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA (2010) Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature 465: 793–797.
  24. 24. Vallinoto AC, Graça ES, Araújo MS, Azevedo VN, Cayres-Vallinoto I, et al. (2010) IFNG +874 T/A polymorphism and cytokine plasma levels are associated with susceptibility to Mycobacterium tuberculosis infection and clinical manifestation of tuberculosis. . Hum. Immunol 71: 692–696.
  25. 25. He J, Wang J, Lei D, Ding S (2010) Analysis of functional SNP in ifng/ifngr1 in Chinese Han population with tuberculosis. Scand J Immunol 71: 452–458.
  26. 26. Stein CM, Zalwango S, Malone LL, Won S, Mayanja-Kizza H, et al. (2008) Genome scan of M. tuberculosis infection and disease in Ugandans. PLoS One 3: e4094.
  27. 27. Liu H, Kang H, Liu R, Chen X, Zhao K (2002) Maximal induction of a subset of interferon target genes requires the chromatin-remodeling activity of the BAF complex. Mol Cell Biol 22: 6471–6479.
  28. 28. Cuddapah S, Cui K, Zhao K (2008) Transcriptional enhancer factor 1 (TEF-1/TEAD1) mediates activation of IFITM3 gene by BRGl. FEBS Lett 582: 391–397.
  29. 29. Li D, Peng Z, Tang H, Wei P, Kong X, et al. (2011) KLF4-mediated negative regulation of IFITM3 expression plays a critical role in colon cancer pathogenesis. Clin Cancer Res 17: 3558–3568.
  30. 30. Friedman RL, Manly SP, McMahon M, Kerr IM, Stark GR (1984) Transcriptional and posttranscriptional regulation of interferon-induced gene expression in human cells. Cell 38: 745–755.
  31. 31. Wu XR, Yin QQ, Jiao AX, Xu BP, Sun L, et al. (2012) Pediatric tuberculosis at Beijing Children's Hospital: 2002–2010. Pediatrics 130: e1433–1440.
  32. 32. The Respiratory Disease Group in Pediatrics Society of Chinese Medical Association (2006) The Editorial Board of Chinese Journal of Pediatrics, Diagnostic standards and therapeutic recommendations for pulmonary tuberculosis in children. Chin J Pediatr 44: 249–251.
  33. 33. Graham SM, Ahmed T, Amanullah F, Browning R, Cardenas V, et al. (2012) Evaluation of tuberculosis diagnostics in children: 1. Proposed clinical case definitions for classification of intrathoracic tuberculosis disease. Consensus from an expert panel. J Infect Dis 205 Suppl 2S199–208.
  34. 34. Li Z, Zhang Z, He Z, Tang W, Li T, et al. (2009) A partition-ligation-combination-subdivision EM algorithm for haplotype inference with multiallelic markers: update of the SHEsis (http://analysis.bio-x.cn). Cell Res. 19: 519–523.
  35. 35. QUANTO 1.1: A computer program for power and sample size calculations for genetic-epidemiology studies. Gene x Environment, Gene x Gene Interaction Home page website. Available: http://hydra.usc.edu/gxe. Accessed 2013 May 30.
  36. 36. Singh SB, Davis AS, Taylor GA, Deretic V (2006) Human IRGM induces autophagy to eliminate intracellular mycobacteria. Science 313: 1438–1441.
  37. 37. Li X, Yang Y, Zhou F, Zhang Y, Lu H, et al. (2011) SLC11A1 (NRAMP1) polymorphisms and tuberculosis susceptibility: updated systematic review and meta-analysis. PLoS One 6: e15831.