Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 1.
Published in final edited form as: Front Biol (Beijing). 2015 Apr 27;10(3):252–261. doi: 10.1007/s11515-015-1358-y

The bacterial and host factors associated with extrapulmonary dissemination of Mycobacterium tuberculosis

Dong Yang 1, Ying Kong 1,
PMCID: PMC4636013  NIHMSID: NIHMS704623  PMID: 26557138

Abstract

With high morbidity and mortality worldwide, tuberculosis (TB) is still an important public health threat. The majority of human TB cases are caused by Mycobacterium tuberculosis. Although pulmonary TB is the most common presentation, M. tuberculosis can disseminate into other organs and causes extrapulmonary TB (EPTB). The dissemination of bacteria from the initial site of infection to other organs can lead to fatal diseases, such as miliary and meningeal TB. Thoroughly understanding the mechanisms and pathways of dissemination would develop therapies to prevent the lethal prognosis of EPTB (miliary and meningeal TB) and vaccines to promote the development of adaptive immunity. This review focuses on risk factors of EPTB, bacterial and host genes involved in EPTB, and potential mechanisms of M. tuberculosis extrapulmonary dissemination.

Keywords: Mycobacterium tuberculosis, extrapulmonary, dissemination, risk factors, bacterial genes, host genes

Introduction

Although tuberculosis (TB) emerged about 15000 to 35000 years ago (Kapur et al., 1994), it is still an important public health threat globally, causing 8 million new cases and 1.3 million deaths each year (WHO 2014). The majority of human TB cases are caused by Mycobacterium tuberculosis, an aerobic bacterium that can persist in host tissues for months to decades without replication, but resumes growth when host immunity wanes. It is estimated that one-third of the worlds population are latently infected with M. tuberculosis (Sudre et al., 1992). Pulmonary TB is the most common presentation, but M. tuberculosis can disseminate into other organs and causes extrapulmonary TB (EPTB). The trafficking of bacteria from the initial site of infection to other organs can lead to fatal diseases, such as miliary and meningeal TB. Extrapulmonary involvement can occur with or without pulmonary infection sites. About 15% reactivated TB from latency occur at extrapulmonary organs without active pulmonary TB (Hopewell, 1994). It has been reported that M. tuberculosis DNA was isolated from extrapulmonary organs during latent infection in human samples (Barrios-Payán et al., 2012). The rate of EPTB development is between 10% and 25% among immunocompetent patients (Weir and Thornton, 1985; Pitchenik et al., 1988; Snider and Roper, 1992; American Thoracic Society, 2000). Frequent sites of extrapulmonary infection include the pleura, lymph nodes, bones and joints, CNS (meninges), larynx, skeleton (particularly the spine), genitourinary tract, eyes, gastrointestinal tract, adrenal gland, and skin. The clinical presentation of EPTB is atypical. Biopsy and/or surgery are required to procure tissue samples for confirmation of EPTB diagnosis. Thoroughly understanding the mechanisms of M. tuberculosis dissemination would help to prevent the lethal prognosis of EPTB and to improve diagnosis, treatment and prevention of EPTB. This review focuses on risk factors of EPTB, bacterial and host genes involved in EPTB, and potential mechanisms of M. tuberculosis caused extrapulmonary dissemination. Although nontuberculosis mycobacteria can cause both pulmonary and extrapulmonary TB (Alvarado-Esquivel et al., 2009; Henkle and Winthrop, 2015), it is out of the scope of this review.

M. tuberculosis infection

M. tuberculosis is a slow-growing facultative intracellular pathogen that can survive and multiply inside macrophages and other mammalian cells. It is transmitted from patients with active pulmonary disease by droplets, which are then inhaled. After an incubation period of 4 to 12 weeks, approximately one third of the individuals exposed become infected (Edwards and Kirkpatrick, 1986). It is the balance between bacterial virulence and the inherent microbicidal ability of the alveolar macrophages that determines whether an inhaled tubercle bacillus can successfully establish infection in the lungs (Edwards and Kirkpatrick, 1986; Dannenberg, 1989). Once inspired into the lungs, the bacilli multiply and cause inflammation, which induces neutrophils and macrophages to migrate to the area of inflammation. After phagocytizing the bacilli, alveolar macrophages are activated to release cytokines, which recruit more macrophages and activated Tcells to control infection (Dannenberg, 1989). Accumulated macrophages at sites of bacterial implantation further differentiate into epithelioid cells that have tightly interdigitated cell membranes in zipper-like arrays linking adjacent cells to form tuberculous granuloma (Adams, 1976; Bouley et al., 2001). Granuloma contains the pathogen, a large population of Tcells, B cells, dendritic cells, neutrophils, and fibroblasts (Flynn and Chan, 2001; Peters and Ernst, 2003). After granuloma formation, M. tuberculosis is maintained and persists within the center of granuloma in a low active and anaerobic state to avoid direct confrontation with the host immune defense (McKinney et al., 2000). Reactivation happens once the balance between bacillary persistence and the immune response gets disturbed due to aging, malnutrition, steroids or HIV infection (Fenton and Vermeulen, 1996; Flynn and Chan, 2001).

Active TB occurs when the host immune response fails to contain the replication of M. tuberculosis associated with initial infection. It is estimated 5%–10% of those infected with M. tuberculosis develop active TB during the first few years following infection. The clinical manifestations of TB are quite variable and depend on host factors such as age, immune status, coexisting diseases, immunization with BCG, and microbial factors such as virulence of the organism and predilection for specific tissues (American Thoracic Society, 2000). Human immunodeficiency virus (HIV) co-infection increases the risk for active disease of TB. Among HIV-infected persons with latent TB infection, the rates of active disease are up to 100 times higher than those for individuals with latent TB infection without co-infection with HIV (Brewer and Heymann, 2005).

The immune response to M. tuberculosis infection is primarily a cell-mediated response with T cells as the main player (Kaufmann, 2002; Boom et al., 2003). CD4+ T cells, mediated by their cytokine production (Cooper et al., 1993; Jouanguy et al., 1996; de Jong et al., 1998), are the most important aspect of the protective response in M. tuberculosis infection (Caruso et al., 1999; van Pinxteren et al., 2000). One of the cytokines produced by CD4+ and CD8+ T cells is gamma interferon (IFN-γ). It can activate antigen-presenting cells, boost expression of major histocompatibility complex (MHC) (Cooper et al., 1993; Jouanguy et al., 1996; de Jong et al., 1998), induce reactive nitrogen derivatives (especially nitric oxide) (Arias et al., 1997; Nathan and Shiloh, 2000; Shiloh and Nathan, 2000), and alternate phagocytic vesicle tracking/control (MacMicking et al., 2003). IFN-γ can also inhibit over-production of other cytokines, such as tumor necrosis factor α (TNF-α) (Rook and Hernandez-Pando, 1996; Bekker et al., 2000). Strategies employed by M. tuberculosis to evoke T helper-1 cell immune response include resisting intracellular killing mechanisms of microphages, and blocking apoptosis of macrophages (Sly et al., 2003) and the macrophages response to IFN-γ (Fortune et al., 2004).

Risk factors of EPTB

HIV infection

Before the HIV pandemic, about 15%–20% of TB cases developed EPTB (Weir and Thornton, 1985; Pitchenik et al., 1988; Snider and Roper, 1992; American Thoracic Society, 2000). In HIV-positive patients, however, EPTB cases increased dramatically to more than 50% of all cases of TB (Theuer et al., 1990; Shafer et al., 1991; Haas and Des Prez, 1994; Antonucci et al., 1995; Lado Lado et al., 1999; Lee et al., 2000; Yang et al., 2004). We found the risk of developing EPTB in HIV positive patients is as high as 5-fold of that in HIV negative patients, after controlling age, race, and gender (Yang et al., 2004). The close association between HIV infection and EPTB is very likely due to deficiency of CD4+ T cells among HIV infected patients. It is well known that HIV targets on CD4+ T cells and causes reduced CD4+ T cells and less cytokine production. CD4+ T-helper cells are major players for controlling M. tuberculosis infection. Among HIV positive patients, the risk of EPTB increases as the CD4+ lymphocyte count declines (Jones et al., 1993). The most common extrapulmonary site in HIV-positive individuals is the lymph node. However, other extrapulmonary sites such as neurological, pleural, pericardial, abdominal involvement have also been described in HIV-positive patients (Raviglione et al., 1992; Barnes and Barrows, 1993; Jones et al., 1993).

Race/ethnicity

Population-based epidemiological studies reported that African Americans tend to have higher proportion of EPTB cases (Farer et al., 1979; Yang et al., 2004; Fiske et al., 2010). In the United States, data from 13 states and two cities reported the rates of EPTB in African Americans is 5 times of that in Whites (6.5 vs 1.3 per 100000 population) (Farer et al., 1979). In a case-control study with over 700 TB patients, we have found the risk of developing EPTB in African Americans is as 2-fold high as in White population, after controlling for other confounding factors, including age, gender and HIV coninfection (Yang et al., 2004). Higher risk of EPTB development in African American population may be related to higher overall TB incidence rates (Centers for Disease Control and Prevention, 2008), higher exposure to TB, lower socioeconomic status, and lack of access to medical care (Rieder et al., 1990). Additionally higher prevalence of HIV infection might lead to higher risk of EPTB in African Americans.

People born in South Asian countries have higher incidence rates of EPTB than other foreign-born patients (Asghar et al., 2008). The mechanism is unknown. It might be due to unidentified host genetic or physiological factors that increase their risks to develop disseminated disease. One possible cause is vitamin D deficiency. Vitamin D deficiency happens more in dark-skinned people (Harris, 2006). The association between vitamin D deficiency and susceptibility to TB has been reported in epidemiologic and laboratory-based studies (Liu et al., 2006; Martineau et al., 2007; Sita-Lumsden et al., 2007). Although there was not significant independent association between the vitamin D receptor (VDR) genotype and TB susceptibility, the combination of TT/Tt genotype of VDR gene and vitamin D deficiency was significantly associated with TB susceptibility (Wilkinson et al., 2000). In clinical studies, vitamin D supplementation was observed to reduce patient pulmonary lesion (Nursyam et al., 2006). It has been reported that 25-hydroxyvitamin D can be activated by 25-hydroxyvitamin D-1α hydroxylase (CYP27B1) into 1,25-dihydroxyvitamin D3. Binding of active vitamin D to its receptor VDR on cell surface increases expressions of β-defensin 2 and cathelicidin (Wang et al., 2004; Gombart et al., 2005), which stimulate autophagy of M. tuberculosis infected phagocytes (Campbell and Spector, 2012), and help to inhibit bacterial replication. Vitamin D was also found to induce IL-1β expression in M. tuberculosis infected macrophages cocultured with human small airway epithelial cells, and to reduce bacterial burden through another antimicrobial peptide, DEFB4/HBD2, generated by the cocultured epithelial cells in response to IL-1β (Verway et al., 2013).

Gender

While men typically have higher overall rates of TB compared with women (Martinez et al., 2000), some studies have shown that among people who develop TB, women are more likely to have EPTB than men (Rieder et al., 1990; Chan-Yeung et al., 2002; Yang et al., 2004; Musellim et al., 2005; Sreeramareddy et al., 2008). A population-based case-control control study in the United States has showed that 16.9% of female patients had extrapulmonary disease compared to 9.3% of male patients (Yang et al., 2004). Similar findings have also been seen in Asian and European populations. (Chan-Yeung et al., 2002; Forssbohm et al., 2008; Zhang et al., 2011; Lin et al., 2013). It is still unclear why females tend to have more EPTB. Hormonal factors, smoking and TB exposure might be the causes of this inequality (Hudelson, 1996; Holmes et al., 1998). Older women are less able to contain bacilli in the lungs due to reduced levels of sex hormones after menopause. The prevalence of smoking is higher in males than females. Smoking is a risk factor for pulmonary TB (Bates et al., 2007; Chiang et al., 2007). Another report has suggested that smoking is associated with relapse of TB and smokers are less likely to have isolated EPTB (Chiang et al., 2007). Other possible factors accounting for the difference are stigma associated with having TB and lack of access to health care, especially for females in resource limited regions (Holmes et al., 1998).

Age

Whether age is an independent risk factor of EPTB is not certain. In the above mentioned population-based case-control study, it has been found that the risk of EPTB among younger than 25 years old is 2-fold of that among older patients (Yang et al., 2004). Other studies from the United States (Gonzalez et al., 2003) and Europe (Cailhol et al., 2005) have also reported that younger age was an independent risk factor for EPTB. However, another study from Turkey (Musellim et al., 2005) has reported that age was not associated with EPTB. These inconsistent findings could be attributed to different prevalence of host-related factors or important co-exposures among the studied populations.

Bacterial genes involved in EPTB

After infection of M. tuberculosis in lungs, whether EPTB occurs is likely determined by interactions between the pathogen and the host immune response. Studies on bacterial virulence genes using animal models have identified several genes that might be related to M. tuberculosis dissemination.

Seven genes whiB1 through whiB7 at separate loci are in the M. tuberculosis genome (Cole et al., 1998; Camus et al., 2002) as orthologs of the whiB gene of Streptomyces coelicolor A3(2), which was annotated as a putative transcription factor, and has been shown to be involved in sporulation (Davis and Chater, 1992). Among them, WhiB4 has been postulated to act as a sensor of oxidative stress. M. tbΔwhiB4 showed a defect in dissemination to guinea-pig spleen, suggesting that whiB4 is essential for successful dissemination. It is likely WhiB4 regulates oxidative stress response to modulate survival in macrophages, and thus helps bacterial dissemination (Chawla et al., 2012).

A locus in M. tuberculosis genome, designated as mel2, plays an important role during persistence in mice (Cirillo et al., 2009). Like the whiB4, the mel2 mutant displays increased susceptibility to reactive oxygen species (ROS) (Cirillo et al., 2009). In aerosol infected mice, the mutant grew normally until the persistent stage, where it did not persist as well as the wild type, resulted in reduced pathology and CFU in spleen at 4 weeks post infection (Cirillo et al., 2009).

The most severe type of EPTB is meningitis, which happens when M. tuberculosis infects the central nervous system (CNS). Bacterial genes required for invasion or survival in mouse CNS were identified by using a pooled defined M. tuberculosis mutants library to intravenously infect mice (Be et al., 2008), including Rv0311, Rv0805, Rv0931c, and Rv0986. Rv0805 and Rv0986 mutants were significantly attenuated on day 1 in brain tissue, in comparison with them in blood, suggesting that they have roles in invasion of the CNS. In addition, Rv0311, Rv0805, and Rv0931c might also play roles in survival in the CNS, because mutants of Rv0311, Rv0805, and Rv0931c were found to be significantly attenuated in brain on day 49, in comparison with them in brain on day 1(Be et al., 2008). Using rabbits infected intrathecally with different M. tuberculosis clinical isolates, the group of rabbits infected with HN878, a M. tuberculosis strain caused 60 cases of TB in Texas from 1995 to 1998 (Manca et al., 2001), was found having the highest bacillary load in the brain and the most severe leukocytosisin cerebrospinal fluid (Tsenova et al., 2005). The higher bacterial load and leukocytosis in CNS caused by HN878 was ascribed to a polyketide synthase (PKS)–derived phenolic glycolipid (PGL), because the PKS genes deleted strain HN878pks1-15∷hyg infected animals showed reduced bacterial load, less severe pathologic changes and attenuated clinical manifestations (Tsenova et al., 2005), and the PGL-deficient mutant of HN878 was found to be more immunogenic and less lethal in infected mice (Reed et al., 2004).

Another region named “igr” for the defect in intracellular growth might play roles in dissemination, intracellular survival, and lipid catabolism (Sassetti and Rubin, 2003; Schnappinger et al., 2003; Rengarajan et al., 2005; Chang et al., 2007). The igr region is composed of 6 genes (Rv3540c– Rv3545c) (Chang et al., 2007). Three of these six open reading frames are lying in the same orientation and annotated as a cytochrome P450 (cyp125), and two acyl-coenzyme A (CoA) dehydrogenases (fadE28 and fadE29); the other three genes are two conserved hypotheticals (Rv3541c and Rv3542c), and one lipid transfer protein (ltp2) (Chang et al., 2007). Aerosol infection of mice showed the strain having a deletion of this region had delayed dissemination to the spleen, and reduced lung pathology (Chang et al., 2007). However, the deletion mutant of this region showed no difference in persistence in comparison with the wild type strain (Chang et al., 2007).

Host genes involved in disseminated TB

After M. tuberculosis infection, bacterial replication and dissemination are first controlled by the host innate immune response, and then by a T cell mediated adaptive immune response. The innate immune response to M. tuberculosis is primarily through macrophages and intracellular signaling pathways. Bacterial antigen pattern recognition receptors, such as Toll-like receptors, are involved in bacterial recognition and macrophage activation. Toll-like receptor 2 (TLR2) has been found to recognize M. tuberculosis and initiate the innate immune response to infection. TLR2 genotype T597C was found associated with TB meningitis in a case-control study (Thuong et al., 2007). The association increased with the severity of neurologic symptoms (Thuong et al., 2007). TNF-α is an important cytokine for controlling M. tuberculosis infection. Individuals treated with the anti-TNF-α agent infliximab were found associated with disseminated TB (Keane et al., 2001). Another anti-TNF-α agent, adalimumab, caused disseminated TB in non-human primate model (Lin et al., 2010).

Besides main factors in innate immunity, cytokines and chemokines playing roles in T cell mediated immunity are also involved in dissemination of M. tuberculosis, including genes encoding IFN-y receptor, IL-12 receptor, and the signal transducer and activator of transcription-1 (STAT-1). A mutation in the IFN-γR1 chain was identified from six children with disseminated environmental mycobacterial infection, suggesting that the IFN-γR gene mutation is associated with mycobacterial dissemination (Levin et al., 1995). Mutations in IL-12 receptor β 1 chain were found from three patients having severe, recurrent, and systemic Mycobacterial and Salmonella infections (de Jong et al., 1998). These patients also had reduced IFN-γ production from NK cells and T cells (de Jong et al., 1998). A genetic polymorphism in the Manose Binding Protein (MBP) encoding gene was associated with TB meningitis in a South African population (Hoal-Van Helden et al., 1999). The MBP B allele (G54D) led to low MBP levels, which provided protection against tuberculous meningitis (Hoal-Van Helden et al., 1999). Genetic polymorphisms in interleukin (IL)-1p/ IL-1R (Wilkinson et al., 1999), IL-10, IFN-γ (Henao et al., 2006), and NRAMP1 (Kim et al., 2003) were associated with pleural TB. In addition, a polymorphism in the P2X7 gene, which encodes a receptor expressed on macrophages, was associated with EPTB (Fernando et al., 2007).

Mechanisms of dissemination

It is well accepted that M. tuberculosis can migrate from the primary infection site, lungs, to the lymphatic system and bloodstream. However, the detailed mechanisms of bacterial dissemination remain unclear. To migrate from lungs to the draining lymph nodes and blood stream, the bacilli must break through alveolar epithelium. So far, some evidences have shown that bacteria inside alveolar macrophages or dendritic cells can be relocated by these professional phagocytes into lymph nodes and blood. Bacteria could also invade and lyse epithelial cells after infecting epithelial cells.

Bacteria relocated by professional phagocytes

One of the ways for M. tuberculosis to infect other organs is to use professional phagocytes as vehicles to approach organs far away from lungs by following blood stream. After phagocytized by professional phagocytes, mycobacteria can survive within phagosomes, the hostile acidic niche, and prevent fusion of phagosome to lysosome (McDonough et al., 1993) by using sulfatides (Goren et al., 1976), by producing ammonia or ammonium chlorides (Gordon et al., 1980; Hart et al., 1983), and by deactivating calmodulin and calmodulin dependent protein kinase 2 (Malik et al., 2001). Once infection is established, the infected macrophages and dendritic cells are surrounded by T cells and B cells to form granulomas. It was a dogma that granuloma helps confine bacterial dissemination. However, studies using zebra fish embryo infected with M. marinum demonstrated that macrophages within granulomas helped mycobacteria disseminate from initial infection sites to distant sites (Clay et al., 2007; Davis and Ramakrishnan, 2009). Early secretory antigenic target 6 kDa (ESAT6) and culture filtrate protein 10 kDa (CFP10) are two proteins encoded by Rv3874 and Rv3875 within the RD1 gene cluster, which is present in M. tuberculosis genome but not in the M. bovis bacillus Calmette-Guérin (BCG) genome (Cole, 2002). In the M. marinum infected zebra fish embryo, RD1 deleted mutant could not disseminate to distant sites (Clay et al., 2007). It suggested that genes in the RD1 region are important for bacterial dissemination by migrating within granulomas. These studies were conducted with M. marinum in immune immature zebra fish model. It remains to test whether these observations hold true in M. tuberculosis infected mature mammal model.

Another professional phagocyte, dendritic cell, also plays important roles in controlling M. tuberculosis infection. Using aerosol infected mouse model and flow cytometry, it has been found that the majority of infected cells in the lungs and mediastinl lymph node were CD11chighCD11bhigh myeloid DCs and recruited macrophages (Wolf et al., 2007). After phagocytizing mycobacteria, dendritic cells present bacterial antigens to T cells to prime IFN-γ producing T cells and to induce cell-mediated response to infection (Tascon et al., 2000). Mannosylated lipoarabinomannan (ManLAM) on mycobacteria cell wall can interact with a c-type lectin receptor on dendritic cells to modify host response to inhibit dendritic cell maturation (Fortune et al., 2004). These investigations suggest M. tuberculosis may utilize macrophages and dendritic cells to traffic to the lymph nodes and blood (Menozzi et al., 1996).

Bacteria invade and lyse epithelial cells

Epithelial cells are the first lining of the alveolus that inhaled M. tuberculosis interacts with. After infecting epithelial cells, M. tuberculosis can replicate and undergo cell-to-cell spreading (Castro-Garza et al., 2002). Some studies suggest that M. tuberculosis can lyse epithelial cells and cause necrosis of epithelial cells (Dobos et al., 2000). Cytotoxicity for epithelial cells is associated with bacterial virulence (McDonough and Kress, 1995). The heparin binding hemagglutinin (HbhA) of M. tuberculosis was found to play important roles in infecting epithelial cells. HbhA was initially identified in M. tuberculosis and M. bovis (BCG) (Menozzi et al., 1996; Menozzi et al., 1998). It is located on the surface of the mycobacterium and mediates binding of the bacillus to epithelial cells and fibroblasts (Menozzi et al., 1996). Mutation of hbhA leaded to reduced adherence of bacteria to epithelial cells but no effect on adherence to macrophages (Pethe et al., 2001). In intranasal infected mice, mutation of hbhA did not affect colonization in lungs, but severely reduced colonization in spleen. However, this discrepancy was not observed when infection is through i.v., suggesting that HbhA is critical for escaping from lung, but not for colonization in extrapulmonary organs (Pethe et al., 2001; Sohn et al., 2011; Lebrun et al., 2012). Overall, these studies advocate that HbhA is important for M. tuberculosis to infect and transcytose epithelial cell layer and to escape from lungs.

Mammalian cell entry (mce) genes were identified when a DNA fragment of M. tuberculosis H37Ra was transformed into E. coli, which enabled this non-pathogenic bacterium to invade a nonphagocytic HeLa cell line. Initially, this gene was named mce (Arruda et al., 1993). Subsequently, other paralogous genes were found in M. tuberculosis H37Rv and these genes are now named mce1A, 2A, 3A, and 4A (Casali and Riley, 2007). These four gene are located in four separated mce operons, mce1 to mce4, with similar organization and a 450 bp core sequence (Kumar et al., 2003). In mice intratracheally infected with mutants of mce 2 or 3, bacterial loads in both lung and spleen are significantly lower than the bacterial loads in the wild type strain; whereas in intraperitonealy infected mice, there were not significantly different bacterial loads in lung and spleen among the mutant and wild type strains infected groups (Gioffré et al., 2005). These results indicate genes in mce 2 and mce 3 operons might be involved in extrapulmonary dissemination. It remains to further characterize the detailed mechanisms and functions of each genes of mce operons.

Although ESAT6 and CFP10 were shown to be important for dissemination through the mechanism of being relocated by professional phagocytes as above discussed, there are evidences showing that they could help bacterial dissemination by lysing epithelial cells. Transposon disruption of the esat6 and cfp10 operon of RD1 showed defect of cytolysis in pneumocytes and macrophages (Hsu et al., 2003). ESAT6 can bind to laminin on the basolateral surface of alveolar epithelial cells and lyse pneumocytes (Kinhikar et al., 2010). In this way, ESAT6 helps bacteria directly disseminate through the alveolar wall.

Summary

Among TB patients, 10%–20% can develop EPTB, which complicates diagnosis and treatments, and thus increases morbidity and mortality of the disease. Although many factors have been reported involved or associated with EPTB development, the detailed mechanisms remain unclear. The phenotype of M. tuberculosis dissemination from initial infection site in the infected host is a sign of failure of containing bacteria by the granuloma. However, since lymph nodes are the critical place for professional phagocytes to present antigens to T cells, and thus to prime T cells, bacterial dissemination into regional lymph nodes also helps to develop protective T cell mediated immune response. There have been many evidences showing both bacterial and host factors play important roles in M. tuberculosis dissemination. As studies on bacterial and host genes related to dissemination move forward, more and more new factors and pathways will be identified and characterized. These results will, without a doubt, broaden and deepen our knowledge of TB pathogenesis, and thus improve our methods of diagnosis, treatment and prevention of TB.

Acknowledgments

Work in the authors' laboratory was supported by the National Heart, Lung, And Blood Institute of the National Institutes of Health under Award Number R21HL115463. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Footnotes

Compliance with ethics guidelines: Dong Yang and Ying Kong declare that they have no conflict of interests. This manuscript is a review article and does not involve a research protocol requiring approval by the relevant institutional review board or ethics committee.

References

  1. Adams DO. The granulomatous inflammatory response. A review. Am J Pathol. 1976;84(1):164–192. [PMC free article] [PubMed] [Google Scholar]
  2. Alvarado-Esquivel C, García-Corral N, Carrero-Dominguez D, Enciso-Moreno JA, Gurrola-Morales T, Portillo-Gómez L, Rossau R, Mijs W. Molecular analysis of Mycobacterium isolates from extrapulmonary specimens obtained from patients in Mexico. BMC Clin Pathol. 2009;9(1):1. doi: 10.1186/1472-6890-9-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. American Thoracic Society, Infectious Diseases Society of America. Diagnostic standards and classification of tuberculosis in adults and children. Am J Respir Crit Care Med. 2000;161(4 Pt 1):1376–1395. doi: 10.1164/ajrccm.161.4.16141. [DOI] [PubMed] [Google Scholar]
  4. Antonucci G, Girardi E, Raviglione MC, Ippolito G. Risk factors for tuberculosis in HIV-infected persons. A prospective cohort study. The Gruppo Italiano di Studio Tubercolosi e AIDS (GISTA) JAMA. 1995;274(2):143–148. doi: 10.1001/jama.274.2.143. [DOI] [PubMed] [Google Scholar]
  5. Arias M, Zabaleta J, Rodríguez JI, Rojas M, París SC, García LF. Failure to induce nitric oxide production by human monocyte-derived macrophages. Manipulation of biochemical pathways. Allergol Immunopathol (Madr) 1997;25(6):280–288. [PubMed] [Google Scholar]
  6. Arruda S, Bomfim G, Knights R, Huima-Byron T, Riley LW. Cloning of an M. tuberculosis DNA fragment associated with entry and survival inside cells. Science. 1993;261(5127):1454–1457. doi: 10.1126/science.8367727. [DOI] [PubMed] [Google Scholar]
  7. Asghar RJ, Pratt RH, Kammerer JS, Navin TR. Tuberculosis in South Asians living in the United States, 1993–2004. Arch Intern Med. 2008;168(9):936–942. doi: 10.1001/archinte.168.9.936. [DOI] [PubMed] [Google Scholar]
  8. Barnes PF, Barrows SA. Tuberculosis in the 1990s. Ann Intern Med. 1993;119(5):400–410. doi: 10.7326/0003-4819-119-5-199309010-00009. [DOI] [PubMed] [Google Scholar]
  9. Barrios-Payán J, Saqui-Salces M, Jeyanathan M, Alcántara-Vazquez A, Castañon-Arreola M, Rook G, Hernandez-Pando R. Extra-pulmonary locations of Mycobacterium tuberculosis DNA during latent infection. J Infect Dis. 2012;206(8):1194–1205. doi: 10.1093/infdis/jis381. [DOI] [PubMed] [Google Scholar]
  10. Bates MN, Khalakdina A, Pai M, Chang L, Lessa F, Smith KR. Risk of tuberculosis from exposure to tobacco smoke: a systematic review and meta-analysis. Arch Intern Med. 2007;167(4):335–342. doi: 10.1001/archinte.167.4.335. [DOI] [PubMed] [Google Scholar]
  11. Be NA, Lamichhane G, Grosset J, Tyagi S, Cheng QJ, Kim KS, Bishai WR, Jain SK. Murine model to study the invasion and survival of Mycobacterium tuberculosis in the central nervous system. J Infect Dis. 2008;198(10):1520–1528. doi: 10.1086/592447. [DOI] [PubMed] [Google Scholar]
  12. Bekker LG, Moreira AL, Bergtold A, Freeman S, Ryffel B, Kaplan G. Immunopathologic effects of tumor necrosis factor alpha in murine mycobacterial infection are dose dependent. Infect Immun. 2000;68(12):6954–6961. doi: 10.1128/iai.68.12.6954-6961.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Boom WH, Canaday DH, Fulton SA, Gehring AJ, Rojas RE, Torres M. Human immunity to M. tuberculosis: T cell subsets and antigen processing. Tuberculosis (Edinb) 2003;83(1-3):98–106. doi: 10.1016/s1472-9792(02)00054-9. [DOI] [PubMed] [Google Scholar]
  14. Bouley DM, Ghori N, Mercer KL, Falkow S, Ramakrishnan L. Dynamic nature of host-pathogen interactions in Mycobacterium marinum granulomas. Infect Immun. 2001;69(12):7820–7831. doi: 10.1128/IAI.69.12.7820-7831.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brewer TF, Heymann SJ. Long time due: reducing tuberculosis mortality in the 21st century. Arch Med Res. 2005;36(6):617–621. doi: 10.1016/j.arcmed.2005.06.002. [DOI] [PubMed] [Google Scholar]
  16. Cailhol J, Decludt B, Che D. Sociodemographic factors that contribute to the development of extrapulmonary tuberculosis were identified. J Clin Epidemiol. 2005;58(10):1066–1071. doi: 10.1016/j.jclinepi.2005.02.023. [DOI] [PubMed] [Google Scholar]
  17. Campbell GR, Spector SA. Vitamin D inhibits human immunodeficiency virus type 1 and Mycobacterium tuberculosis infection in macrophages through the induction of autophagy. PLoS Pathog. 2012;8(5):e1002689. doi: 10.1371/journal.ppat.1002689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Camus JC, Pryor MJ, Médigue C, Cole ST. Re-annotation of the genome sequence of Mycobacterium tuberculosis H37Rv. Microbiology. 2002;148(Pt 10):2967–2973. doi: 10.1099/00221287-148-10-2967. [DOI] [PubMed] [Google Scholar]
  19. Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, Flynn JL. Mice deficient in CD4 T cells have only transiently diminished levels of IFN-gamma, yet succumb to tuberculosis. J Immunol. 1999;162(9):5407–5416. [PubMed] [Google Scholar]
  20. Casali N, Riley LW. A phylogenomic analysis of the Actinomycetales mce operons. BMC Genomics. 2007;8(1):60. doi: 10.1186/1471-2164-8-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Castro-Garza J, King CH, Swords WE, Quinn FD. Demonstration of spread by Mycobacterium tuberculosis bacilli in A549 epithelial cell monolayers. FEMS Microbiol Lett. 2002;212(2):145–149. doi: 10.1111/j.1574-6968.2002.tb11258.x. [DOI] [PubMed] [Google Scholar]
  22. Centers for Disease Control and Prevention (CDC) Trends in tuberculosis—United States, 2007. MMWR Morb Mortal Wkly Rep. 2008;57(11):281–285. [PubMed] [Google Scholar]
  23. Chan-Yeung M, Noertjojo K, Chan SL, Tam CM. Sex differences in tuberculosis in Hong Kong. Int J Tuberc Lung Dis. 2002;6(1):11–18. [PubMed] [Google Scholar]
  24. Chang JC, Harik NS, Liao RP, Sherman DR. Identification of Mycobacterial genes that alter growth and pathology in macrophages and in mice. J Infect Dis. 2007;196(5):788–795. doi: 10.1086/520089. [DOI] [PubMed] [Google Scholar]
  25. Chawla M, Parikh P, Saxena A, Munshi M, Mehta M, Mai D, Srivastava AK, Narasimhulu KV, Redding KE, Vashi N, Kumar D, Steyn AJ, Singh A. Mycobacterium tuberculosis WhiB4 regulates oxidative stress response to modulate survival and dissemination in vivo. Mol Microbiol. 2012;85(6):1148–1165. doi: 10.1111/j.1365-2958.2012.08165.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chiang CY, Slama K, Enarson DA. Associations between tobacco and tuberculosis. Int J Tuberc Lung Dis. 2007;11(3):258–262. [PubMed] [Google Scholar]
  27. Cirillo SL, Subbian S, Chen B, Weisbrod TR, Jacobs WR, Jr, Cirillo JD. Protection of Mycobacterium tuberculosis from reactive oxygen species conferred by the mel2 locus impacts persistence and dissemination. Infect Immun. 2009;77(6):2557–2567. doi: 10.1128/IAI.01481-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Clay H, Davis JM, Beery D, Huttenlocher A, Lyons SE, Ramakrishnan L. Dichotomous role of the macrophage in early Mycobacterium marinum infection of the zebrafish. Cell Host Microbe. 2007;2(1):29–39. doi: 10.1016/j.chom.2007.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cole ST. Comparative and functional genomics of the Mycobacterium tuberculosis complex. Microbiology. 2002;148(Pt 10):2919–2928. doi: 10.1099/00221287-148-10-2919. [DOI] [PubMed] [Google Scholar]
  30. Cole ST, Brosch R, Parkhill J, Garnier T, Churcher C, Harris D, Gordon SV, Eiglmeier K, Gas S, Barry CE, 3rd, Tekaia F, Badcock K, Basham D, Brown D, Chillingworth T, Connor R, Davies R, Devlin K, Feltwell T, Gentles S, Hamlin N, Holroyd S, Hornsby T, Jagels K, Krogh A, McLean J, Moule S, Murphy L, Oliver K, Osborne J, Quail MA, Rajandream MA, Rogers J, Rutter S, Seeger K, Skelton J, Squares R, Squares S, Sulston JE, Taylor K, Whitehead S, Barrell BG. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature. 1998;393(6685):537–544. doi: 10.1038/31159. [DOI] [PubMed] [Google Scholar]
  31. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme IM. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med. 1993;178(6):2243–2247. doi: 10.1084/jem.178.6.2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dannenberg AM., Jr Immune mechanisms in the pathogenesis of pulmonary tuberculosis. Rev Infect Dis. 1989;11(Suppl 2):S369–S378. doi: 10.1093/clinids/11.supplement_2.s369. [DOI] [PubMed] [Google Scholar]
  33. Davis JM, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009;136(1):37–49. doi: 10.1016/j.cell.2008.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Davis NK, Chater KF. The Streptomyces coelicolor whiB gene encodes a small transcription factor-like protein dispensable for growth but essential for sporulation. Mol Gen Genet. 1992;232(3):351–358. doi: 10.1007/BF00266237. [DOI] [PubMed] [Google Scholar]
  35. de Jong R, Altare F, Haagen IA, Elferink DG, Boer T, van Breda Vriesman PJ, Kabel PJ, Draaisma JM, van Dissel JT, Kroon FP, Casanova JL, Ottenhoff TH. Severe mycobacterial and Salmonella infections in interleukin-12 receptor-deficient patients. Science. 1998;280(5368):1435–1438. doi: 10.1126/science.280.5368.1435. [DOI] [PubMed] [Google Scholar]
  36. Dobos KM, Spotts EA, Quinn FD, King CH. Necrosis of lung epithelial cells during infection with Mycobacterium tuberculosis is preceded by cell permeation. Infect Immun. 2000;68(11):6300–6310. doi: 10.1128/iai.68.11.6300-6310.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Edwards D, Kirkpatrick CH. The immunology of mycobacterial diseases. Am Rev Respir Dis. 1986;134(5):1062–1071. doi: 10.1164/arrd.1986.134.5.1062. [DOI] [PubMed] [Google Scholar]
  38. Farer LS, Lowell AM, Meador MP. Extrapulmonary tuberculosis in the United States. Am J Epidemiol. 1979;109(2):205–217. doi: 10.1093/oxfordjournals.aje.a112675. [DOI] [PubMed] [Google Scholar]
  39. Fenton MJ, Vermeulen MW. Immunopathology of tuberculosis: roles of macrophages and monocytes. Infect Immun. 1996;64(3):683–690. doi: 10.1128/iai.64.3.683-690.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Fernando SL, Saunders BM, Sluyter R, Skarratt KK, Goldberg H, Marks GB, Wiley JS, Britton WJ. A polymorphism in the P2X7 gene increases susceptibility to extrapulmonary tuberculosis. Am J Respir Crit Care Med. 2007;175(4):360–366. doi: 10.1164/rccm.200607-970OC. [DOI] [PubMed] [Google Scholar]
  41. Fiske CT, Griffin MR, Erin H, Warkentin J, Lisa K, Arbogast PG, Sterling TR. Black race, sex, and extrapulmonary tuberculosis risk: an observational study. BMC Infect Dis. 2010;10(1):16. doi: 10.1186/1471-2334-10-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol. 2001;19(1):93–129. doi: 10.1146/annurev.immunol.19.1.93. [DOI] [PubMed] [Google Scholar]
  43. Forssbohm M, Zwahlen M, Loddenkemper R, Rieder HL. Demographic characteristics of patients with extrapulmonary tuberculosis in Germany. Eur Respir J. 2008;31(1):99–105. doi: 10.1183/09031936.00020607. [DOI] [PubMed] [Google Scholar]
  44. Fortune SM, Solache A, Jaeger A, Hill PJ, Belisle JT, Bloom BR, Rubin EJ, Ernst JD. Mycobacterium tuberculosis inhibits macrophage responses to IFN-gamma through myeloid differentiation factor 88-dependent and-independent mechanisms. J Immunol. 2004;172(10):6272–6280. doi: 10.4049/jimmunol.172.10.6272. [DOI] [PubMed] [Google Scholar]
  45. Gioffré A, Infante E, Aguilar D, Santangelo MP, Klepp L, Amadio A, Meikle V, Etchechoury I, Romano MI, Cataldi A, Hernández RP, Bigi F. Mutation in mce operons attenuates Mycobacterium tuberculosis virulence. Microbes Infect. 2005;7(3):325–334. doi: 10.1016/j.micinf.2004.11.007. [DOI] [PubMed] [Google Scholar]
  46. Gombart AF, Borregaard N, Koeffler HP. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J. 2005;19(9):1067–1077. doi: 10.1096/fj.04-3284com. [DOI] [PubMed] [Google Scholar]
  47. Gonzalez OY, Adams G, Teeter LD, Bui TT, Musser JM, Graviss EA. Extra-pulmonary manifestations in a large metropolitan area with a low incidence of tuberculosis. Int J Tuberc Lung Dis. 2003;7(12):1178–1185. [PubMed] [Google Scholar]
  48. Gordon AH, Hart PD, Young MR. Ammonia inhibits phagosome-lysosome fusion in macrophages. Nature. 1980;286(5768):79–80. doi: 10.1038/286079a0. [DOI] [PubMed] [Google Scholar]
  49. Goren MB, D'Arcy Hart P, Young MR, Armstrong JA. Prevention of phagosome-lysosome fusion in cultured macrophages by sulfatides of Mycobacterium tuberculosis. Proc Natl Acad Sci USA. 1976;73(7):2510–2514. doi: 10.1073/pnas.73.7.2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Haas DW, Des Prez RM. Tuberculosis and acquired immunodeficiency syndrome: a historical perspective on recent developments. Am J Med. 1994;96(5):439–450. doi: 10.1016/0002-9343(94)90171-6. [DOI] [PubMed] [Google Scholar]
  51. Harris SS. Vitamin D and African Americans. J Nutr. 2006;136(4):1126–1129. doi: 10.1093/jn/136.4.1126. [DOI] [PubMed] [Google Scholar]
  52. Hart PD, Young MR, Jordan MM, Perkins WJ, Geisow MJ. Chemical inhibitors of phagosome-lysosome fusion in cultured macrophages also inhibit saltatory lysosomal movements. A combined microscopic and computer study. J Exp Med. 1983;158(2):477–492. doi: 10.1084/jem.158.2.477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Henao MI, Montes C, París SC, García LF. Cytokine gene polymorphisms in Colombian patients with different clinical presentations of tuberculosis. Tuberculosis (Edinb) 2006;86(1):11–19. doi: 10.1016/j.tube.2005.03.001. [DOI] [PubMed] [Google Scholar]
  54. Henkle E, Winthrop KL. Nontuberculous mycobacteria infections in immunosuppressed hosts. Clin Chest Med. 2015;36(1):91–99. doi: 10.1016/j.ccm.2014.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hoal-Van Helden EG, Epstein J, Victor TC, Hon D, Lewis LA, Beyers N, Zurakowski D, Ezekowitz AB, Van Helden PD. Mannosebinding protein B allele confers protection against tuberculous meningitis. Pediatr Res. 1999;45(4 Pt 1):459–464. doi: 10.1203/00006450-199904010-00002. [DOI] [PubMed] [Google Scholar]
  56. Holmes CB, Hausler H, Nunn P. A review of sex differences in the epidemiology of tuberculosis. Int J Tuberc Lung Dis. 1998;2(2):96–104. [PubMed] [Google Scholar]
  57. Hopewell P. Overview of Clinical Tuberculosis. In: Barry B, editor. Tuberculosis: Pathogenesis, Protection and Control. ASM Press; Washington, DC: 1994. pp. 25–46. [Google Scholar]
  58. Hsu T, Hingley-Wilson SM, Chen B, Chen M, Dai AZ, Morin PM, Marks CB, Padiyar J, Goulding C, Gingery M, Eisenberg D, Russell RG, Derrick SC, Collins FM, Morris SL, King CH, Jacobs WR., Jr The primary mechanism of attenuation of bacillus Calmette-Guerin is a loss of secreted lytic function required for invasion of lung interstitial tissue. Proc Natl Acad Sci USA. 2003;100(21):12420–12425. doi: 10.1073/pnas.1635213100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hudelson P. Gender differentials in tuberculosis: the role of socio-economic and cultural factors. Tuber Lung Dis. 1996;77(5):391–400. doi: 10.1016/s0962-8479(96)90110-0. [DOI] [PubMed] [Google Scholar]
  60. Jones BE, Young SM, Antoniskis D, Davidson PT, Kramer F, Barnes PF. Relationship of the manifestations of tuberculosis to CD4 cell counts in patients with human immunodeficiency virus infection. Am Rev Respir Dis. 1993;148(5):1292–1297. doi: 10.1164/ajrccm/148.5.1292. [DOI] [PubMed] [Google Scholar]
  61. Jouanguy E, Altare F, Lamhamedi S, Revy P, Emile JF, Newport M, Levin M, Blanche S, Seboun E, Fischer A, Casanova JL. Interferon-gamma-receptor deficiency in an infant with fatal bacille Calmette-Guérin infection. N Engl J Med. 1996;335(26):1956–1961. doi: 10.1056/NEJM199612263352604. [DOI] [PubMed] [Google Scholar]
  62. Kapur V, Whittam TS, Musser JM. Is Mycobacterium tuberculosis 15,000 years old? J Infect Dis. 1994;170(5):1348–1349. doi: 10.1093/infdis/170.5.1348. [DOI] [PubMed] [Google Scholar]
  63. Kaufmann SH. Protection against tuberculosis: cytokines, T cells, and macrophages. Ann Rheum Dis. 2002;61(Suppl 2):ii54–ii58. doi: 10.1136/ard.61.suppl_2.ii54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Keane J, Gershon S, Wise RP, Mirabile-Levens E, Kasznica J, Schwieterman WD, Siegel JN, Braun MM. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med. 2001;345(15):1098–1104. doi: 10.1056/NEJMoa011110. [DOI] [PubMed] [Google Scholar]
  65. Kim JH, Lee SY, Lee SH, Sin C, Shim JJ, In KH, Yoo SH, Kang KH. NRAMP1 genetic polymorphisms as a risk factor of tuberculous pleurisy. Int J Tuberc Lung Dis. 2003;7(4):370–375. [PubMed] [Google Scholar]
  66. Kinhikar AG, Verma I, Chandra D, Singh KK, Weldingh K, Andersen P, Hsu T, Jacobs WR, Jr, Laal S. Potential role for ESAT6 in dissemination of M. tuberculosis via human lung epithelial cells. Mol Microbiol. 2010;75(1):92–106. doi: 10.1111/j.1365-2958.2009.06959.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kumar A, Bose M, Brahmachari V. Analysis of expression profile of mammalian cell entry (mce) operons of Mycobacterium tuberculosis. Infect Immun. 2003;71(10):6083–6087. doi: 10.1128/IAI.71.10.6083-6087.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Lado Lado FL, Barrio Gómez E, Carballo Arceo E, Cabarcos Ortíz de Barrón A, Lado FL, Barrio Gómez E. Clinical presentation of tuberculosis and the degree of immunodeficiency in patients with HIV infection. Scand J Infect Dis. 1999;31(4):387–391. doi: 10.1080/00365549950163842. [DOI] [PubMed] [Google Scholar]
  69. Lebrun P, Raze D, Fritzinger B, Wieruszeski JM, Biet F, Dose A, Carpentier M, Schwarzer D, Allain F, Lippens G, Locht C. Differential contribution of the repeats to heparin binding of HBHA, a major adhesin of Mycobacterium tuberculosis. PLoS ONE. 2012;7(3):e32421. doi: 10.1371/journal.pone.0032421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Lee MP, Chan JW, Ng KK, Li PC. Clinical manifestations of tuberculosis in HIV-infected patients. Respirology. 2000;5(4):423–426. [PubMed] [Google Scholar]
  71. Levin M, Newport MJ, D'Souza S, Kalabalikis P, Brown IN, Lenicker HM, Agius PV, Davies EG, Thrasher A, Klein N, et al. Familial disseminated atypical mycobacterial infection in childhood: a human mycobacterial susceptibility gene? Lancet. 1995;345(8942):79–83. doi: 10.1016/s0140-6736(95)90059-4. [DOI] [PubMed] [Google Scholar]
  72. Lin CY, Chen TC, Lu PL, Lai CC, Yang YH, Lin WR, Huang PM, Chen YH. Effects of gender and age on development of concurrent extrapulmonary tuberculosis in patients with pulmonary tuberculosis: a population based study. PLoS ONE. 2013;8(5):e63936. doi: 10.1371/journal.pone.0063936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lin PL, Myers A, Smith L, Bigbee C, Bigbee M, Fuhrman C, Grieser H, Chiosea I, Voitenek NN, Capuano SV, Klein E, Flynn JL. Tumor necrosis factor neutralization results in disseminated disease in acute and latent Mycobacterium tuberculosis infection with normal granuloma structure in a cynomolgus macaque model. Arthritis Rheum. 2010;62(2):340–350. doi: 10.1002/art.27271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, Ochoa MT, Schauber J, Wu K, Meinken C, Kamen DL, Wagner M, Bals R, Steinmeyer A, Zügel U, Gallo RL, Eisenberg D, Hewison M, Hollis BW, Adams JS, Bloom BR, Modlin RL. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770–1773. doi: 10.1126/science.1123933. [DOI] [PubMed] [Google Scholar]
  75. MacMicking JD, Taylor GA, McKinney JD. Immune control of tuberculosis by IFN-gamma-inducible LRG-47. Science. 2003;302(5645):654–659. doi: 10.1126/science.1088063. [DOI] [PubMed] [Google Scholar]
  76. Malik ZA, Iyer SS, Kusner DJ. Mycobacterium tuberculosis phagosomes exhibit altered calmodulin-dependent signal transduction: contribution to inhibition of phagosome-lysosome fusion and intracellular survival in human macrophages. J Immunol. 2001;166(5):3392–3401. doi: 10.4049/jimmunol.166.5.3392. [DOI] [PubMed] [Google Scholar]
  77. Manca C, Tsenova L, Bergtold A, Freeman S, Tovey M, Musser JM, Barry CE, 3rd, Freedman VH, Kaplan G. Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-alpha /beta. Proc Natl Acad Sci USA. 2001;98(10):5752–5757. doi: 10.1073/pnas.091096998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Martineau AR, Wilkinson RJ, Wilkinson KA, Newton SM, Kampmann B, Hall BM, Packe GE, Davidson RN, Eldridge SM, Maunsell ZJ, Rainbow SJ, Berry JL, Griffiths CJ. A single dose of vitamin D enhances immunity to mycobacteria. Am J Respir Crit Care Med. 2007;176(2):208–213. doi: 10.1164/rccm.200701-007OC. [DOI] [PubMed] [Google Scholar]
  79. Martinez AN, Rhee JT, Small PM, Behr MA. Sex differences in the epidemiology of tuberculosis in San Francisco. Int J Tuberc Lung Dis. 2000;4(1):26–31. [PubMed] [Google Scholar]
  80. McDonough KA, Kress Y. Cytotoxicity for lung epithelial cells is a virulence-associated phenotype of Mycobacterium tuberculosis. Infect Immun. 1995;63(12):4802–4811. doi: 10.1128/iai.63.12.4802-4811.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. McDonough KA, Kress Y, Bloom BR. Pathogenesis of tuberculosis: interaction of Mycobacterium tuberculosis with macrophages. Infect Immun. 1993;61(7):2763–2773. doi: 10.1128/iai.61.7.2763-2773.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. McKinney JD, Höner zu Bentrup K, Muñoz-Elías EJ, Miczak A, Chen B, Chan WT, Swenson D, Sacchettini JC, Jacobs WR, Jr, Russell DG. Persistence of Mycobacterium tuberculosis in macrophages and mice requires the glyoxylate shunt enzyme isocitrate lyase. Nature. 2000;406(6797):735–738. doi: 10.1038/35021074. [DOI] [PubMed] [Google Scholar]
  83. Menozzi FD, Bischoff R, Fort E, Brennan MJ, Locht C. Molecular characterization of the mycobacterial heparin-binding hemagglutinin, a mycobacterial adhesin. Proc Natl Acad Sci USA. 1998;95(21):12625–12630. doi: 10.1073/pnas.95.21.12625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Menozzi FD, Rouse JH, Alavi M, Laude-Sharp M, Muller J, Bischoff R, Brennan MJ, Locht C. Identification of a heparin-binding hemagglutinin present in mycobacteria. J Exp Med. 1996;184(3):993–1001. doi: 10.1084/jem.184.3.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Musellim B, Erturan S, Sonmez Duman E, Ongen G. Comparison of extra-pulmonary and pulmonary tuberculosis cases: factors influencing the site of reactivation. Int J Tuberc Lung Dis. 2005;9(11):1220–1223. [PubMed] [Google Scholar]
  86. Nathan C, Shiloh MU. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. Proc Natl Acad Sci USA. 2000;97(16):8841–8848. doi: 10.1073/pnas.97.16.8841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Nursyam EW, Amin Z, Rumende CM. The effect of vitamin D as supplementary treatment in patients with moderately advanced pulmonary tuberculous lesion. Acta Med Indones. 2006;38(1):3–5. [PubMed] [Google Scholar]
  88. Peters W, Ernst JD. Mechanisms of cell recruitment in the immune response to Mycobacterium tuberculosis. Microbes Infect. 2003;5(2):151–158. doi: 10.1016/s1286-4579(02)00082-5. [DOI] [PubMed] [Google Scholar]
  89. Pethe K, Alonso S, Biet F, Delogu G, Brennan MJ, Locht C, Menozzi FD. The heparin-binding haemagglutinin of M. tuberculosis is required for extrapulmonary dissemination. Nature. 2001;412(6843):190–194. doi: 10.1038/35084083. [DOI] [PubMed] [Google Scholar]
  90. Pitchenik AE, Fertel D, Bloch AB. Mycobacterial disease: epidemiology, diagnosis, treatment, and prevention. Clin Chest Med. 1988;9(3):425–441. [PubMed] [Google Scholar]
  91. Raviglione MC, Narain JP, Kochi A. HIV-associated tuberculosis in developing countries: clinical features, diagnosis, and treatment. Bull World Health Organ. 1992;70(4):515–526. [PMC free article] [PubMed] [Google Scholar]
  92. Reed MB, Domenech P, Manca C, Su H, Barczak AK, Kreiswirth BN, Kaplan G, Barry CE., 3rd A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature. 2004;431(7004):84–87. doi: 10.1038/nature02837. [DOI] [PubMed] [Google Scholar]
  93. Rengarajan J, Bloom BR, Rubin EJ. Genome-wide requirements for Mycobacterium tuberculosis adaptation and survival in macrophages. Proc Natl Acad Sci USA. 2005;102(23):8327–8332. doi: 10.1073/pnas.0503272102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rieder HL, Snider DE, Jr, Cauthen GM. Extrapulmonary tuberculosis in the United States. Am Rev Respir Dis. 1990;141(2):347–351. doi: 10.1164/ajrccm/141.2.347. [DOI] [PubMed] [Google Scholar]
  95. Rook GA, Hernandez-Pando R. The pathogenesis of tuberculosis. Annu Rev Microbiol. 1996;50(1):259–284. doi: 10.1146/annurev.micro.50.1.259. [DOI] [PubMed] [Google Scholar]
  96. Sassetti CM, Rubin EJ. Genetic requirements for mycobacterial survival during infection. Proc Natl Acad Sci USA. 2003;100(22):12989–12994. doi: 10.1073/pnas.2134250100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Schnappinger D, Ehrt S, Voskuil MI, Liu Y, Mangan JA, Monahan IM, Dolganov G, Efron B, Butcher PD, Nathan C, Schoolnik GK. Transcriptional adaptation of Mycobacterium tuberculosis within Macrophages: Insights into the phagosomal environment. J Exp Med. 2003;198(5):693–704. doi: 10.1084/jem.20030846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Shafer RW, Kim DS, Weiss JP, Quale JM. Extrapulmonary tuberculosis in patients with human immunodeficiency virus infection. Medicine (Baltimore) 1991;70(6):384–397. doi: 10.1097/00005792-199111000-00004. [DOI] [PubMed] [Google Scholar]
  99. Shiloh MU, Nathan CF. Reactive nitrogen intermediates and the pathogenesis of Salmonella and Mycobacteria. Curr Opin Microbiol. 2000;3(1):35–42. doi: 10.1016/s1369-5274(99)00048-x. [DOI] [PubMed] [Google Scholar]
  100. Sita-Lumsden A, Lapthorn G, Swaminathan R, Milburn HJ. Reactivation of tuberculosis and vitamin D deficiency: the contribution of diet and exposure to sunlight. Thorax. 2007;62(11):1003–1007. doi: 10.1136/thx.2006.070060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Sly LM, Hingley-Wilson SM, Reiner NE, McMaster WR. Survival of Mycobacterium tuberculosis in host macrophages involves resistance to apoptosis dependent upon induction of antiapoptotic Bcl-2 family member Mcl-1. J Immunol. 2003;170(1):430–437. doi: 10.4049/jimmunol.170.1.430. [DOI] [PubMed] [Google Scholar]
  102. Snider DE, Jr, Roper WL. The new tuberculosis. N Engl J Med. 1992;326(10):703–705. doi: 10.1056/NEJM199203053261011. [DOI] [PubMed] [Google Scholar]
  103. Sohn H, Kim JS, Shin SJ, Kim K, Won CJ, Kim WS, Min KN, Choi HG, Lee JC, Park JK, Kim HJ. Targeting of Mycobacterium tuberculosis heparin-binding hemagglutinin to mitochondria in macrophages. PLoS Pathog. 2011;7(12):e1002435. doi: 10.1371/journal.ppat.1002435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Sreeramareddy CT, Panduru KV, Verma SC, Joshi HS, Bates MN. Comparison of pulmonary and extrapulmonary tuberculosis in Nepal- a hospital-based retrospective study. BMC Infect Dis. 2008;8(1):8. doi: 10.1186/1471-2334-8-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sudre P, ten Dam G, Kochi A. Tuberculosis: a global overview of the situation today. Bull World Health Organ. 1992;70(2):149–159. [PMC free article] [PubMed] [Google Scholar]
  106. Tascon RE, Soares CS, Ragno S, Stavropoulos E, Hirst EM, Colston MJ. Mycobacterium tuberculosis-activated dendritic cells induce protective immunity in mice. Immunology. 2000;99(3):473–480. doi: 10.1046/j.1365-2567.2000.00963.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Theuer CP, Hopewell PC, Elias D, Schecter GF, Rutherford GW, Chaisson RE. Human immunodeficiency virus infection in tuberculosis patients. J Infect Dis. 1990;162(1):8–12. doi: 10.1093/infdis/162.1.8. [DOI] [PubMed] [Google Scholar]
  108. Thuong NT, Hawn TR, Thwaites GE, Chau TT, Lan NT, Quy HT, Hieu NT, Aderem A, Hien TT, Farrar JJ, Dunstan SJ. A polymorphism in human TLR2 is associated with increased susceptibility to tuberculous meningitis. Genes Immun. 2007;8(5):422–428. doi: 10.1038/sj.gene.6364405. [DOI] [PubMed] [Google Scholar]
  109. Tsenova L, Ellison E, Harbacheuski R, Moreira AL, Kurepina N, Reed MB, Mathema B, Barry CE, 3rd, Kaplan G. Virulence of selected Mycobacterium tuberculosis clinical isolates in the rabbit model of meningitis is dependent on phenolic glycolipid produced by the bacilli. J Infect Dis. 2005;192(1):98–106. doi: 10.1086/430614. [DOI] [PubMed] [Google Scholar]
  110. van Pinxteren LA, Cassidy JP, Smedegaard BH, Agger EM, Andersen P. Control of latent Mycobacterium tuberculosis infection is dependent on CD8 T cells. Eur J Immunol. 2000;30(12):3689–3698. doi: 10.1002/1521-4141(200012)30:12<3689::AID-IMMU3689>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  111. Verway M, Bouttier M, Wang TT, Carrier M, Calderon M, An BS, Devemy E, McIntosh F, Divangahi M, Behr MA, White JH. Vitamin D induces interleukin-1β expression: paracrine macrophage epithelial signaling controls M. tuberculosis infection. PLoS Pathog. 2013;9(6):e1003407. doi: 10.1371/journal.ppat.1003407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, Tavera-Mendoza L, Lin R, Hanrahan JW, Mader S, White JH. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol. 2004;173(5):2909–2912. doi: 10.4049/jimmunol.173.5.2909. [DOI] [PubMed] [Google Scholar]
  113. Weir MR, Thornton GF. Extrapulmonary tuberculosis. Experience of a community hospital and review of the literature. Am J Med. 1985;79(4):467–478. doi: 10.1016/0002-9343(85)90034-8. [DOI] [PubMed] [Google Scholar]
  114. WHO. Global tuberculosis control 2013 2014 [Google Scholar]
  115. Wilkinson RJ, Llewelyn M, Toossi Z, Patel P, Pasvol G, Lalvani A, Wright D, Latif M, Davidson RN. Influence of vitamin D deficiency and vitamin D receptor polymorphisms on tuberculosis among Gujarati Asians in west London: a case-control study. Lancet. 2000;355(9204):618–621. doi: 10.1016/S0140-6736(99)02301-6. [DOI] [PubMed] [Google Scholar]
  116. Wilkinson RJ, Patel P, Llewelyn M, Hirsch CS, Pasvol G, Snounou G, Davidson RN, Toossi Z. Influence of polymorphism in the genes for the interleukin (IL)-1 receptor antagonist and IL-1beta on tuberculosis. J Exp Med. 1999;189(12):1863–1874. doi: 10.1084/jem.189.12.1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Wolf AJ, Linas B, Trevejo-Nuñez GJ, Kincaid E, Tamura T, Takatsu K, Ernst JD. Mycobacterium tuberculosis infects dendritic cells with high frequency and impairs their function in vivo. J Immunol. 2007;179(4):2509–2519. doi: 10.4049/jimmunol.179.4.2509. [DOI] [PubMed] [Google Scholar]
  118. Yang Z, Kong Y, Wilson F, Foxman B, Fowler AH, Marrs CF, Cave MD, Bates JH. Identification of risk factors for extrapulmonary tuberculosis. Clin Infect Dis. 2004;38(2):199–205. doi: 10.1086/380644. [DOI] [PubMed] [Google Scholar]
  119. Zhang X, Andersen AB, Lillebaek T, Kamper-Jørgensen Z, Thomsen VO, Ladefoged K, Marrs CF, Zhang L, Yang Z. Effect of sex, age, and race on the clinical presentation of tuberculosis: a 15-year population-based study. Am J Trop Med Hyg. 2011;85(2):285–290. doi: 10.4269/ajtmh.2011.10-0630. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES