Skip to main content
Advertisement
  • Loading metrics

Experimental study of tuberculosis: From animal models to complex cell systems and organoids

  • Kaori L. Fonseca,

    Affiliations i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal

  • Pedro N. S. Rodrigues,

    Affiliations i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal

  • I. Anna S. Olsson,

    Affiliations i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal

  • Margarida Saraiva

    margarida.saraiva@ibmc.up.pt

    Affiliations i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal

Abstract

Tuberculosis (TB) is a devastating disease to mankind that has killed more people than any other infectious disease. Despite many efforts and successes from the scientific and health communities, the prospect of TB elimination remains distant. On the one hand, sustainable public health programs with affordable and broad implementation of anti-TB measures are needed. On the other hand, achieving TB elimination requires critical advances in three areas: vaccination, diagnosis, and treatment. It is also well accepted that succeeding in advancing these areas requires a deeper knowledge of host—pathogen interactions during infection, and for that, better experimental models are needed. Here, we review the potential and limitations of different experimental approaches used in TB research, focusing on animal and human-based cell culture models. We highlight the most recent advances in developing in vitro 3D models and introduce the potential of lung organoids as a new tool to study Mycobacterium tuberculosis infection.

Author summary

Tuberculosis (TB) is the number 1 killer in the world due to a bacterial infection. The study of this disease through clinical and epidemiological data and through the use of different experimental models has provided important knowledge on the role of the immune response generated during infection. This is critical for the development of novel vaccines and therapeutic strategies. However, in spite of the advances made, it is well accepted that better models are needed to study TB. This review discusses the different models used to study TB, highlighting the advantages and disadvantages of the available animal and cellular models and introducing recently developed state-of-the-art approaches based on human-based cell culture systems. These new advances are integrated in a road map for future study of TB, converging for the potential of lung organoids in TB research.

General introduction

Tuberculosis (TB) kills over 1.8 million people every year and thus remains the leading cause of death by an infectious agent [1]. Additionally, TB afflicts over 10.4 million new individuals per year and is estimated to exist in a latent form in nearly 2 billion people worldwide [1]. In addition to the human toll, TB imposes a significant economic burden, corresponding to 0.52% of the global gross national product, with a cost of over 500 million euros per year in the European Union alone [2]. Tackling TB is therefore a matter of urgency, as reflected in the current WHO End TB Strategy, which targets a 90% reduction in the incidence of TB to less than 100 cases per million people by 2035 [3]. Achieving this target requires a much quicker decline in TB incidence, from the current annual reduction of 2% to a 20% decrease per year [4, 5]. For this, 3 areas in TB research are generally accepted as critical: development of novel vaccines, improved diagnostic tools, and better treatment options [5, 6]. Succeeding in advancing these areas requires fresh approaches and ways of thinking, notably the development of better experimental models to study TB [7]. In this review, we discuss the different experimental approaches used in TB research, from in vivo models to human-based cell culture ones (Table 1). We also propose a road map of the available experimental approaches to study TB and of alternatives that are envisaged in a near future (Fig 1). We highlight the most recent advances in developing in vitro 3D models and introduce the potential of lung organoids as a new tool to study host—pathogen interactions during Mycobacterium tuberculosis infection. The development of such models requires a deep understanding of the disease pathogenesis and of the immune players, which are not the focus of this review and have been extensively reviewed elsewhere [810].

thumbnail
Table 1. Experimental models for the study of tuberculosis (TB).

https://doi.org/10.1371/journal.ppat.1006421.t001

thumbnail
Fig 1. An integrative view on the experimental models for tuberculosis (TB) research.

Questions arising from clinical, genetic, and epidemiological data on TB are addressed using a variety of experimental approaches. The traditional approach has used a combination of 2D culture systems and animal models. The recent development of 3D cell culture systems composed by multiple cell types provides in vitro models with a level of complexity previously only available in vivo. The generation of lung-on-chip cultures and the possibility of generating lung organoids from healthy or patient donors may in future offer experimental systems closer to the human pathophysiology. Abbreviations: ESC, embryonic stem cells; iPSC, induced pluripotent stem cells; PSC, pluripotent stem cells.

https://doi.org/10.1371/journal.ppat.1006421.g001

In vivo models in TB research

Several animal models are used in TB research (Fig 2), ranging from zebrafish to nonhuman primates (NHPs) [11, 12]. Mice are preferred model animals for a number of practical reasons, such as availability of immunological-based tools for mice, the existence of genetically modified mouse strains, and the small size and cost-effectiveness of maintaining mice in the laboratory [1315]. Whereas many important aspects of the immune system are indeed conserved, there are also important differences that hamper the use of the mouse model of infection in our understanding of TB pathogenesis. The mouse is not a natural host for M. tuberculosis, and lung cavitation, a key characteristic for the disease transmission in humans [16], is not observed for the 2 most-used mouse strains (Balb/c and BL6) [13]. Necrotizing responses to M. tuberculosis occur in other mouse strains [17], indicating the impact of genetic variability on the outcome of infection. A recent study illustrates this fact by demonstrating that the susceptibility to TB infection and the efficacy of Bacillus Calmette-Guerin (BCG) vaccination varied greatly when genetically different mouse strains were used [18]. It is thus not surprising that, depending on the mouse strain used, different studies report different data. Furthermore, variability in the reported results is enhanced by different experimental end points used [19, 20]. The route and dose of M. tuberculosis administration and the mouse microbiome are also thought to contribute to variable findings.

thumbnail
Fig 2. Proportion of different animal models in TB research.

Pie chart illustrating the percentage of publications for each of the most commonly used animal models in TB research. Results from a Pubmed search performed on 9 February 2017 using the following key words: “mouse AND tuberculosis,” “guinea pig AND tuberculosis,” “rabbit AND tuberculosis,” “non-human primate AND tuberculosis,” and “zebra fish AND tuberculosis.” Percentages were calculated based on the total number of publications for all animal models.

https://doi.org/10.1371/journal.ppat.1006421.g002

Since currently used mouse models fail to fully reflect human immunity to TB, several studies were performed using humanized mice. Humanized mice can be generated through the reconstitution of immunocompromised mice with human hematopoietic cells of different origins [21]. Infection of humanized mice with M. tuberculosis reproduced important hallmark features of human TB disease pathology, such as the formation of organized granulomatous lesions, caseous necrosis, and bronchial obstruction [22, 23]. However, abnormal T-cell responses and an impaired bacterial control were also observed [23]. In line with this, humanized mice generated by engraftment of human leukocyte antigen (HLA)-restricted cells showed partial function of innate and adaptive immune systems, culminating in antigen-specific T-cell responses to mycobacterial infection but also in lack of protection [24]. Other approaches consist in infecting transgenic mice expressing human-specific molecules such as, for example, the human cluster of differentiation group 1 CD1, which allows for the study of a humanized immune system using the mouse model of infection [25]. In all, humanized mice are a good tool to study TB, being particularly relevant for the study of HIV/TB, as recently shown [26]. However, this model requires further improvement to reach its full potential for TB research.

To address some of these limitations, other animal models have been used. For example, guinea pigs and rabbits may be considered better models to study the humanlike granuloma formation, a hallmark of M. tuberculosis infection in the lung [14, 27, 28], although they still fail to display other characteristics of the human disease. Additionally, they are much more difficult to maintain in the lab and a lot less immunology tools are available for these 2 species, which greatly limits their use. Infection of zebrafish (Danio rerio) embryos with the natural fish pathogen M. marinum is also used as a model for the study of granuloma formation [2931]. Several similarities were found in the cellular and molecular events presiding M. marinum and M. tuberculosis infections [3234], despite the many differences between these 2 diseases. Research on zebrafish embryos benefits from the similarities between M. marinum and M. tuberculosis, i.e., from the optical transparency of the embryos, which facilitates the use of advanced imaging techniques, and from the easy genetic manipulation of zebrafish, which allows for deep mechanistic molecular studies. Because zebrafish embryos lack a fully developed immune system, the study of later stages of infection requires the use of adult fish, thus abrogating the advantages of using embryos. Furthermore, the physiological differences between zebrafish and humans are enormous, which inevitably imposes some limitations to the use of this model. As for the other animal models, specific facilities for housing zebrafish are required. NHPs are so far considered as the best animal model for TB research [35, 36], as the disease pathogenesis parallels that observed in humans [37]. NHPs present lung cavitation [38]; show a spectrum of disease overlapping that of humans, namely, with the establishment of latent TB infection [38]; display a susceptibility to TB in the presence of comorbidities such as HIV and anti—tumor necrosis factor (TNF) treatment similar to that reported in humans [39, 40]; and present a transcriptomic signature of disease comparable to the human one [41]. However, the ethical, practical, and economic problems that are inherent to NHP research [36, 42], exacerbated when the animals are made to develop a potentially fatal infection, hinder the generalized use of this animal model, which in fact accounts for only 1% of the papers published in TB (Fig 2). In conclusion, important advances in our understanding of TB have been made through the use of different animal models. However, in addition to each model’s specific limitations, all animal-model research into human diseases is ultimately restricted by the need to translate findings across species. This calls for the wider use of human-based models to complement and reduce the use of experimental in vivo research.

Human-based in vitro models in TB research

Owing to the central role of the macrophage as host and effector cell during M. tuberculosis infection [43, 44], many studies have been centered in macrophage cell cultures. In terms of human-based systems, monocyte-derived macrophages are the most widely used culture. Among these is the human monocytic leukemia cell line, THP-1, which is easy to culture, yielding a nearly unlimited amount of cells for experimental purposes. THP-1 cells are typically differentiated to macrophages through the stimulation with phorbol 12-myristate 13-acetate (PMA) for 3 days, although different protocols are found in the literature [45, 46], which may contribute to some variable findings. Macrophages can alternatively be freshly derived by extracting and culturing human peripheral blood mononuclear cells (PBMCs) in the presence of differentiating factors, namely, granulocyte-macrophage colony stimulating factor (GM-CSF) or macrophage colony stimulating factor (M-CSF) [47], or of human serum [48]. In these cases, the macrophages are of primary origin, but because of the in vitro differentiation process, their properties are most likely different from tissue-resident cells. Although alveolar macrophages would be ideally used, access to these cells is a costly procedure that requires lengthy ethical approvals, which limits their use. In vivo, M. tuberculosis is found in foamy macrophages. These cells result from pathogen-induced dysregulation of host lipid synthesis and sequestration and play a key role in both sustaining persistent bacteria and contributing to the tissue pathology [49]. Therefore, in vitro differentiation of foamy macrophages is an excellent tool for the study of macrophage-pathogen interactions. A protocol to convert cultured macrophages (THP-1 or primary) into foamy cells has been developed by incubating these cells under hypoxia [50]. Other alternatives for the differentiation of foamy cells include the exposure of cell cultures to palmitic acid, oleic acid, or lipoproteins [51] or to surfactant lipids [52].

Given the importance of working with primary, unmanipulated cells, many studies have been performed using freshly isolated human PBMCs [53]. Human PBMCs are easily accessible, cost-effective, and readily infected with M. tuberculosis, responding to the infection with the production of relevant immune mediators such as TNF and other interleukins as well as chemokines [53]. Furthermore, the PBMC response captures interactions between different immune cell types, such as monocytes, T cells, and B cells, which are in fact interacting during natural immune responses. However, these cells still differ from the tissue-resident ones and when used in in vitro cultures lack the environmental stimuli that ultimately shape cellular responses to infection. In addition to the standard monolayer cultures, PBMCs have been used to develop in vitro models of human mycobacterial granulomas. In 1 study [54], a sequential recruitment of human monocytes and lymphocytes towards mycobacterial antigen-coated artificial beads or live mycobacteria was observed. This recruitment culminated with the formation of a cellular structure reminiscent of natural mycobacterial granulomas in terms of morphology and cell differentiation [54]. This or similar/improved models have been used in several studies [5557]. A different approach based on the culture of human PBMCs in a collagen matrix with a low dose of M. tuberculosis was used to develop an in vitro model of human TB granuloma with dormant bacteria [58]. This model recapitulated important characteristics of the mycobacterial granuloma, such as the aggregation of lymphocytes surrounding infected macrophages, the formation of multinucleated giant cells, the presence of secreted cytokines and chemokines in the culture supernatants, and the reactivation of M. tuberculosis upon immune suppression caused by TNF blockade [58]. These models offer the possibility of studying the infection by M. tuberculosis in a more physiological environment, resembling the structure of the infected human tissue. They constitute valuable approaches for the study of cell—cell interactions, cell differentiation, and bacterial control.

To further reflect the complex environment and structure of the human lung, a growing body of studies are resorting to the use of new technologies in the tissue-engineering field to advance human-based TB research models into the 3D era (Fig 3) [59]. Tissue bilayer systems consisting of epithelial and endothelial cell layers were initially developed to study the early events of alveolar infection [60, 61]. More recently, through the use of these systems, microfold (M) cells were shown to play a critical role in translocating M. tuberculosis to initiate lung infection [62]. A study combining lung-derived epithelial cells and fibroblasts with peripheral blood primary macrophages reported the establishment of a lung tissue model that upon infection led to the clustering of macrophages reminiscent of early TB granuloma formation [63]. Similarly, another report showed the implementation of an in vitro human 3D lung tissue model to study M. tuberculosis infection that allowed the analysis of human granuloma formation and resembled some features of TB [64].

thumbnail
Fig 3. Use of 3D systems in TB research.

Graph illustrating the increasing number of publications using 3D models for TB research between January 2000 and February 2017, every 3 years. Results are from a Pubmed search performed on 14 February 2017 using the keywords “3D models AND tuberculosis”.

https://doi.org/10.1371/journal.ppat.1006421.g003

A novel bioengineering approach utilized bioelectrospray technology to generate microspheres of M. tuberculosis–infected human PBMCs in a 3D extracellular matrix [59, 65]. This model takes advantage of the high throughput potential of the bioelectrospray system and allows the interrogation of host—pathogen interactions in 3D in the context of an extracellular matrix [59, 66]. When combined with a microfluidic system to enable pharmacokinetic modeling, this model also showed great potential to monitor the efficacy of new antibiotic regimens or anti–M. tuberculosis drugs [65]. Although these experimental systems facilitate the discovery of the interactions between mycobacteria and host cells in a more physiological environment, they still bear some limitations, namely, the lack of vasculature and absence of other immune cells (e.g., neutrophils) that play a role in the multifaceted response in TB infection. Also, not all the models include epithelial and stromal cells, which are known to play important roles during infection [67, 68]. Finally, the spatial organization of the lung is mostly lost, and so is the role of the anatomical constraints during infection. The advances made in the development of all these models will certainly contribute to moving the field forward into novel strategies that overcome current limitations. In this context, other 3D and tissue-chip models are being explored.

Organoids as infection models

Organoids are in vitro 3D cell cultures generated from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), or adult stem cells (aSCs) that functionally and structurally mimic the organ they model [69, 70]. This technology is emerging as a promising tool to study organ development and disease “in a dish” [69, 70]. The potential of organoids to study infectious processes has been increasingly demonstrated in many original papers and recently reviewed by Mills and Estes [71], with most examples coming from human gastric [72], brain [73, 74], and gut [75, 76] organoids. So far, lung organoids have not been explored as a model to study infection.

Lung organoids in TB research

Human lung organoids have been generated through different technologies [77]. The most advanced studies involve the differentiation of human embryonic stem cells into endoderm cells, anterior foregut endoderm cells, lung progenitor cells, and, finally, various types of airway epithelial cells. If this procedure is performed in a 3D structure, a human lung organoid is formed, as initially described by Dye et al. [78] and Konishi et al. [79]. These relatively immature organoids may be transplanted into mice to complete their differentiation in an in vivo environment, into adultlike airways [80].

Despite some limitations, lung organoids recapitulate important features of the lung, such as heterogeneous cell composition, spatial organization, and retention of a stem cell population harboring the capacity for both self-renewal and differentiation [70]. There is increasing evidence that human lung organoids may be used to investigate the cellular and molecular pathways implicated in lung development and lung diseases as well as screening platforms for drugs directed at respiratory diseases [77]. At the disease level, the application of lung organoids to cancer development, cystic fibrosis, and infection is envisaged although still is unexplored in TB research. The obvious advantage of lung organoids over 2D and 3D cultures relies on their spatial organization and heterogeneity of the cellular components. As compared to the animal model, infection of lung organoids allows the inclusion of very early time points, which are difficult to follow in in vivo infections, whilst at the same time overcoming species differences and reducing the use of animals in research. Thus, as the lung organoid technology stands, human-derived lung organoids could be explored to study the early events of infection, namely, the initial interactions of M. tuberculosis with the lung epithelium [67]. Of the aforementioned experimental models, both 2D and 3D cultures based on PBMCs may also be explored to investigate the early immune events during infection, although to a lesser complexity than organoids.

Although there are indeed exciting perspectives for the use of lung organoids as a model for TB research, some important challenges remain before they can be more systematically used as experimental models. Chief among these is the introduction of immune cells in the structure of lung organoids. Only then will lung organoids cover the complexity of immune response and of the stromal-immune cells’ cross talk upon in vitro infection. Also, the introduction of the vasculature would be an important improvement to create a more dynamic model in which the microenvironment of an airway could be experimentally controlled. This dynamic lung organoid would be an interesting model for drug screening. In this context, microfluidic cell culture devices called “organs on a chip” have also generated airway epithelium from human adult airway cells grown on an air—liquid interface platform [81, 82]. Another important step forward would be the development of lung organoids from iPSCs instead of ESCs, as this will offer the possibility of including in the disease modelling individual variability, either genetic or caused by extrinsic conditions. In the context of TB research, this would allow for the study of host–M. tuberculosis–microenvironment interactions at an individual level by infecting lung organoids generated from individuals with HIV or diabetes versus controls or from smokers versus nonsmokers. This would be of utmost importance as the molecular mechanisms underlying the impact of comorbidities and life habits on the course of infection remain incompletely understood. Additionally, comorbidities are very difficult to incorporate in the other complex experimental system—the animal. Generation of personalized lung organoids would also open new avenues for the study of individual responses to therapies and thus for the implementation of personalized medicine.

Conclusions

TB remains a devastating disease to mankind and a huge challenge for the scientific community. From many epidemiological studies, it is clear that the progression of the disease is highly related to the host immune status, and as such, a deep understanding of the immune response to M. tuberculosis is critical for the development of novel preventive and therapeutic strategies. However, the lack of experimental systems that parallel the complexity of the human disease remains a major gap hindering the in-depth study of the immune response in TB. Critical species differences mean that the widely used animal models only partly recapitulate the human disease. NHP models are the most representative ones but bear high operational and maintenance costs. Traditional human cellular systems overcome the interspecies translation problem but are limited by their low level of complexity and the abnormal characteristics of cell lines. Recent development of human-based tissue models is promising real alternatives for the experimental study of human TB. State-of-the art in vitro models have now incorporated several important characteristics of “real-life” tissues, namely, the presence of different cell types and of the extracellular matrix. Technological advances coupled to these models allow for the experimental manipulation of different variables, which is critical in studies of host—pathogen interactions or in drug-screening processes. A key next step will be to introduce in these models the anatomical constraint associated with the lung tissue. Albeit at very early days, lung organoids hold a great promise here. The road from lung organoids to complete lungs “in a dish” is still a long one, but creating a lung structure composed of different stromal cells and coupled with a competent immune system would unquestionably provide a major leap forward in TB research. Being able to use as starting points cells from different individuals (TB patients or latently infected people with different genetic backgrounds and comorbidities) would constitute a revolutionary way of studying TB. This would open many new avenues to investigate long-standing questions and put us in a privileged position to effectively tackle TB.

In sum, recent advances in tissue engineering and future steps in this area will certainly play an important role in the development of new tools for the study of infectious diseases. Such tools hold the potential to replace some animal experiments and overall lead to a reduction of the number of animals used in TB research. Most importantly, these tools will allow for a series of key questions to be answered in a more precise way by including individual variability at the single-cell and tissue levels.

References

  1. 1. WHO. Global Tuberculosis Report. 2016.
  2. 2. Diel R, Rutz S, Castell S, Schaberg T. Tuberculosis: cost of illness in Germany. Eur Respir J. 2012;40[1]:143–51. pmid:22267754
  3. 3. Uplekar M, Weil D, Lonnroth K, Jaramillo E, Lienhardt C, Dias HM, et al. WHO's new End TB Strategy. The Lancet. 2015;385[9979]:1799–801.
  4. 4. Dye C, Glaziou P, Floyd K, Raviglione M. Prospects for tuberculosis elimination. Annu Rev Public Health. 2013;34:271–86. pmid:23244049
  5. 5. Lienhardt C, Lonnroth K, Menzies D, Balasegaram M, Chakaya J, Cobelens F, et al. Translational Research for Tuberculosis Elimination: Priorities, Challenges, and Actions. PLoS Med. 2016;13[3]:e1001965.
  6. 6. Pai M, Behr MA, Dowdy D, Dheda K, Divangahi M, Boehme CC, et al. Tuberculosis. Nat Rev Dis Primers. 2016;2:16076. pmid:27784885
  7. 7. Karp CL, Wilson CB, Stuart LM. Tuberculosis vaccines: barriers and prospects on the quest for a transformative tool. Immunol Rev. 2015;264[1]:363–81. pmid:25703572
  8. 8. O'Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, Berry MP. The immune response in tuberculosis. Annu Rev Immunol. 2013;31:475–527. pmid:23516984
  9. 9. Orme IM, Robinson RT, Cooper AM. The balance between protective and pathogenic immune responses in the TB-infected lung. Nat Immunol. 2015;16[1]:57–63. pmid:25521685
  10. 10. Dorhoi A, Kaufmann SH. Pathology and immune reactivity: understanding multidimensionality in pulmonary tuberculosis. Semin Immunopathol. 2016;38[2]:153–66. pmid:26438324
  11. 11. McMurray DN. Disease model: pulmonary tuberculosis. Trends in molecular medicine. 2001;7[3]:135–7. pmid:11286786
  12. 12. Myllymaki H, Niskanen M, Oksanen KE, Ramet M. Animal models in tuberculosis research—where is the beef? Expert Opin Drug Discov. 2015;10[8]:1–13.
  13. 13. Orme IM. The mouse as a useful model of tuberculosis. Tuberculosis. 2003;83[1–3]:112–5. pmid:12758199
  14. 14. Gupta UD, Katoch VM. Animal models of tuberculosis. Tuberculosis [Edinb]. 2005;85[5–6]:277–93.
  15. 15. Cooper AM. Mouse Model of Tuberculosis. Cold Spring Harb Perspect Med. 2015;5[2]:a018556.
  16. 16. Vilaplana C, Cardona PJ. The lack of a big picture in tuberculosis: the clinical point of view, the problems of experimental modeling and immunomodulation. The factors we should consider when designing novel treatment strategies. Front Microbiol. 2014;5:55. pmid:24592258
  17. 17. Kramnik I, Beamer G. Mouse models of human TB pathology: roles in the analysis of necrosis and the development of host-directed therapies. Semin Immunopathol. 2016;38[2]:221–37. pmid:26542392
  18. 18. Smith CM, Proulx MK, Olive AJ, Laddy D, Mishra BB, Moss C, et al. Tuberculosis Susceptibility and Vaccine Protection Are Independently Controlled by Host Genotype. MBio. 2016;7[5].
  19. 19. Franco NH, Correia-Neves M, Olsson IA. Animal welfare in studies on murine tuberculosis: assessing progress over a 12-year period and the need for further improvement. PLoS ONE. 2012;7[10]:e47723. pmid:23110093
  20. 20. Franco NH, Correia-Neves M, Olsson IA. How "humane" is your endpoint? Refining the science-driven approach for termination of animal studies of chronic infection. PLoS Pathog. 2012;8[1]:e1002399. pmid:22275862
  21. 21. Legrand N, Weijer K, Spits H. Experimental Models to Study Development and Function of the Human Immune System In Vivo. The Journal of Immunology. 2006;176[4]:2053–8. pmid:16455958
  22. 22. Calderon VE, Valbuena G, Goez Y, Judy BM, Huante MB, Sutjita P, et al. A Humanized Mouse Model of Tuberculosis. PLoS ONE. 2013;8[5]:e63331. pmid:23691024
  23. 23. Heuts F, Gavier-Widén D, Carow B, Juarez J, Wigzell H, Rottenberga ME. CD4+ cell-dependent granuloma formation in humanized mice infected with mycobacteria. PNAS. 2013;110[16]:6482–7 pmid:23559373
  24. 24. Lee J, Brehm MA, Greiner D, Shultz LD, Kornfeld H. Engrafted human cells generate adaptive immune responses to Mycobacterium bovis BCG infection in humanized mice. BMC Immunology. 2013;14[53].
  25. 25. Zhao J, Siddiqui S, Shang S, Bian Y, Bagchi S, He Y, et al. Mycolic acid-specific T cells protect against Mycobacterium tuberculosis infection in a humanized transgenic mouse model. Elife. 2015;4.
  26. 26. Nusbaum RJ, Calderon VE, Huante MB, Sutjita P, Vijayakumar S, Lancaster KL, et al. Pulmonary Tuberculosis in Humanized Mice Infected with HIV-1. Sci Rep. 2016;6:21522. pmid:26908312
  27. 27. Arrazuria R, Juste RA, Elguezabal N. Mycobacterial Infections in Rabbits: From the Wild to the Laboratory. Transbound Emerg Dis. 2016.
  28. 28. Clark S, Hall Y, Williams A. Animal models of tuberculosis: Guinea pigs. Cold Spring Harb Perspect Med. 2014;5[5]:a018572. pmid:25524720
  29. 29. Berg RD, Ramakrishnan L. Insights into tuberculosis from the zebrafish model. Trends Mol Med. 2012;18[12]:689–90. pmid:23084762
  30. 30. Myllymaki H, Bauerlein CA, Ramet M. The Zebrafish Breathes New Life into the Study of Tuberculosis. Front Immunol. 2016;7:196. pmid:27242801
  31. 31. Meijer AH. Protection and pathology in TB: learning from the zebrafish model. Semin Immunopathol. 2016;38[2]:261–73.
  32. 32. Ramakrishnan L. The zebrafish guide to tuberculosis immunity and treatment. Cold Spring Harb Symp Quant Biol. 2013;78:179–92. pmid:24643219
  33. 33. Tobin DM, Vary JC Jr., Ray JP, Walsh GS, Dunstan SJ, Bang ND, et al. The lta4h locus modulates susceptibility to mycobacterial infection in zebrafish and humans. Cell. 2010;140[5]:717–30. pmid:20211140
  34. 34. Cronan MR, Beerman RW, Rosenberg AF, Saelens JW, Johnson MG, Oehlers SH, et al. Macrophage Epithelial Reprogramming Underlies Mycobacterial Granuloma Formation and Promotes Infection. Immunity. 2016;45[4]:861–76. pmid:27760340
  35. 35. Flynn JL, Capuano SV, Croix D, Pawar S, Myers A, Zinovik A, et al. Non-human primates: a model for tuberculosis research. Tuberculosis. 2003;83[1–3]:116–8. pmid:12758200
  36. 36. Scanga CA, Flynn JL. Modeling tuberculosis in nonhuman primates. Cold Spring Harb Perspect Med. 2014;4[12]:a018564. pmid:25213189
  37. 37. Flynn JL, Gideon HP, Mattila JT, Lin PL. Immunology studies in non-human primate models of tuberculosis. Immunol Rev. 2015;264:60–73. pmid:25703552
  38. 38. Capuano SV, Croix DA, Pawar S, Zinovik A, Myers A, Lin PL, et al. Experimental Mycobacterium tuberculosis Infection of Cynomolgus Macaques Closely Resembles the Various Manifestations of Human M. tuberculosis Infection. Infect Immun. 2003;71[10]:5831–44. pmid:14500505
  39. 39. Mehra S, Golden NA, Dutta NK, Midkiff CC, Alvarez X, Doyle LA, et al. Reactivation of latent tuberculosis in rhesus macaques by coinfection with simian immunodeficiency virus. J Med Primatol. 2011;40[4]:233–43. pmid:21781131
  40. 40. Lin PL, Myers A, Smith L, Bigbee C, Bigbee M, Fuhrman C, et al. 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]:340–50. pmid:20112395
  41. 41. Gideon HP, Skinner JA, Baldwin N, Flynn JL, Lin PL. Early Whole Blood Transcriptional Signatures Are Associated with Severity of Lung Inflammation in Cynomolgus Macaques with Mycobacterium tuberculosis Infection. J Immunol. 2016;197[12]:4817–28. pmid:27837110
  42. 42. Pena JC, Ho WZ. Monkey models of tuberculosis: lessons learned. Infect Immun. 2015;83[3]:852–62. pmid:25547788
  43. 43. Mayer-Barber KD, Barber DL. Innate and Adaptive Cellular Immune Responses to Mycobacterium tuberculosis Infection. Cold Spring Harb Perspect Med. 2015;5[12].
  44. 44. Tan S, Russel DG. Trans-species communication in the Mycobacterium tuberculosis-infected macrophage. Immunol Rev. 2015;264:233–48. pmid:25703563
  45. 45. Theus SA, Cave MD, Eisenach KD. Activated THP-1 Cells: an Attractive Model for the Assessment of Intracellular Growth Rates of Mycobacterium tuberculosis Isolates. Infect Immun. 2004;72[2]:1169–73. pmid:14742569
  46. 46. Estrella JL, Kan-Sutton C, Gong X, Rajagopalan M, Lewis DE, Hunter RL, et al. A Novel in vitro Human Macrophage Model to Study the Persistence of Mycobacterium tuberculosis Using Vitamin D[3] and Retinoic Acid Activated THP-1 Macrophages. Front Microbiol. 2011;2:67. pmid:21747789
  47. 47. Portevin D, Gagneux S, Comas I, Young D. Human macrophage responses to clinical isolates from the Mycobacterium tuberculosis complex discriminate between ancient and modern lineages. PLoS Pathog. 2011;7[3]:e1001307. pmid:21408618
  48. 48. Vogt G, Nathan C. In vitro differentiation of human macrophages with enhanced antimycobacterial activity. J Clin Invest. 2011;121[10]:3889–901. pmid:21911939
  49. 49. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat Immunol. 2009;10[9]:943–8. pmid:19692995
  50. 50. Daniel J, Maamar H, Deb C, Sirakova TD, Kolattukudy PE. Mycobacterium tuberculosis uses host triacylglycerol to accumulate lipid droplets and acquires a dormancy-like phenotype in lipid-loaded macrophages. PLoS Pathog. 2011;7[6]:e1002093. pmid:21731490
  51. 51. Santucci P, Bouzid F, Smichi N, Poncin I, Kremer L, De Chastellier C, et al. Experimental Models of Foamy Macrophages and Approaches for Dissecting the Mechanisms of Lipid Accumulation and Consumption during Dormancy and Reactivation of Tuberculosis. Front Cell Infect Microbiol. 2016;6:122. pmid:27774438
  52. 52. Dodd CE, Pyle CJ, Glowinski R, Rajaram MV, Schlesinger LS. CD36-Mediated Uptake of Surfactant Lipids by Human Macrophages Promotes Intracellular Growth of Mycobacterium tuberculosis. J Immunol. 2016;197[12]:4727–35. pmid:27913648
  53. 53. Wang C, Peyron P, Mestre O, Kaplan G, van Soolingen D, Gao Q, et al. Innate immune response to Mycobacterium tuberculosis Beijing and other genotypes. PLoS ONE. 2010;5[10]:e13594. pmid:21049036
  54. 54. Puissegur MP, Botanch C, Duteyrat JL, Delsol G, Caratero C, Altare F. An in vitro dual model of mycobacterial granulomas to investigate the molecular interactions between mycobacteria and human host cells. Cell Microbiol. 2004;6[5]:423–33. pmid:15056213
  55. 55. Peyron P, Vaubourgeix J, Poquet Y, Levillain F, Botanch C, Bardou F, et al. Foamy macrophages from tuberculous patients' granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence. PLoS Pathog. 2008;4[11]:e1000204. pmid:19002241
  56. 56. Guirado E, Mbawuike U, Keiser TL, Arcos J, Azad AK, Wang SH, et al. Characterization of host and microbial determinants in individuals with latent tuberculosis infection using a human granuloma model. MBio. 2015;6[1]:e02537–14. pmid:25691598
  57. 57. Yamashiro LH, Eto C, Soncini M, Horewicz V, Garcia M, Schlindwein AD, et al. Isoniazid-induced control of Mycobacterium tuberculosis by primary human cells requires interleukin-1 receptor and tumor necrosis factor. Eur J Immunol. 2016;46[8]:1936–47. pmid:27230303
  58. 58. Kapoor N, Pawar S, Sirakova TD, Deb C, Warren WL, Kolattukudy PE. Human granuloma in vitro model, for TB dormancy and resuscitation. PLoS ONE. 2013;8[1]:e53657. pmid:23308269
  59. 59. Tezera LB, Bielecka MK, Chancellor A, Reichmann MT, Shammari BA, Brace P, et al. Dissection of the host-pathogen interaction in human tuberculosis using a bioengineered 3-dimensional model. Elife. 2017;6:e21283. pmid:28063256
  60. 60. Bermudez LE. The Efficiency of the Translocation of Mycobacterium tuberculosis across a Bilayer of Epithelial and Endothelial Cells as a Model of the Alveolar Wall Is a Consequence of Transport within Mononuclear Phagocytes and Invasion of Alveolar Epithelial Cells. Infection and Immunity. 2002;70[1]:140–6. pmid:11748175
  61. 61. Birkness KA, Deslauriers M, Bartlett JH, White EH, King CH, Quinn FD. An In Vitro Tissue Culture Bilayer Model To Examine Early Events in Mycobacterium tuberculosis Infection. Infect Immun. 1999;67[2]:653–8. pmid:9916072
  62. 62. Nair VR, Franco LH, Zacharia VM, Khan HS, Stamm CE, You W, et al. Microfold Cells Actively Translocate Mycobacterium tuberculosis to Initiate Infection. Cell Rep. 2016;16[5]:1253–8. pmid:27452467
  63. 63. Parasa VR, Rahman MJ, Ngyuen Hoang AT, Svensson M, Brighenti S, Lerm M. Modeling Mycobacterium tuberculosis early granuloma formation in experimental human lung tissue. Dis Model Mech. 2014;7[2]:281–8. pmid:24203885
  64. 64. Braian C, Svensson M, Brighenti S, Lerm M, Parasa VR. A 3D Human Lung Tissue Model for Functional Studies on Mycobacterium tuberculosis Infection. J Vis Exp. 2015[104].
  65. 65. Bielecka MK, Tezera LB, Zmijan R, Drobniewski F, Zhang X, Jayasinghe S, et al. A Bioengineered Three-Dimensional Cell Culture Platform Integrated with Microfluidics To Address Antimicrobial Resistance in Tuberculosis. MBio. 2017;8[1].
  66. 66. Benmerzoug S, Quesniaux VF. Bioengineered 3D Models for Studying Human Cell-Tuberculosis Interactions. Trends Microbiol. 2017;25[4]:245–6. pmid:28284875
  67. 67. Scordo JM, Knoell DL, Torrelles JB. Alveolar Epithelial Cells in Mycobacterium tuberculosis Infection: Active Players or Innocent Bystanders? J Innate Immun. 2016;8[1]:3–14. pmid:26384325
  68. 68. Parida SK, Madansein R, Singh N, Padayatchi N, Master I, Naidu K, et al. Cellular therapy in tuberculosis. Int J Infect Dis. 2015;32:32–8. pmid:25809753
  69. 69. Clevers H. Modeling Development and Disease with Organoids. Cell. 2016;165[7]:1586–97. pmid:27315476
  70. 70. Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol. 2016;18[3]:246–54. pmid:26911908
  71. 71. Mills M, Estes MK. Physiologically relevant human tissue models for infectious diseases. Drug Discov Today. 2016.
  72. 72. McCracken KW, Cata EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 2014;516[7531]:400–4. pmid:25363776
  73. 73. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501[7467]:373–9. pmid:23995685
  74. 74. Ming GL, Tang H, Song H. Advances in Zika Virus Research: Stem Cell Models, Challenges, and Opportunities. Cell Stem Cell. 2016;19[6]:690–702. pmid:27912090
  75. 75. Leslie JL, Huang S, Opp JS, Nagy MS, Kobayashi M, Young VB, et al. Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun. 2015;83[1]:138–45. pmid:25312952
  76. 76. Finkbeiner SR, Zeng XL, Utama B, Atmar RL, Shroyer NF, Estes MK. Stem cell-derived human intestinal organoids as an infection model for rotaviruses. MBio. 2012;3[4]:e00159–12. pmid:22761392
  77. 77. Nadkarni RR, Abed S, Draper JS. Organoids as a model system for studying human lung development and disease. Biochem Biophys Res Commun. 2016;473[3]:675–82. pmid:26721435
  78. 78. Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, et al. In vitro generation of human pluripotent stem cell derived lung organoids. Elife. 2015;4.
  79. 79. Konishi S, Gotoh S, Tateishi K, Yamamoto Y, Korogi Y, Nagasaki T, et al. Directed Induction of Functional Multi-ciliated Cells in Proximal Airway Epithelial Spheroids from Human Pluripotent Stem Cells. Stem Cell Reports. 2016;6[1]:18–25. pmid:26724905
  80. 80. Dye BR, Dedhia PH, Miller AJ, Nagy MS, White ES, Shea LD, et al. A bioengineered niche promotes in vivo engraftment and maturation of pluripotent stem cell derived human lung organoids. Elife. 2016;5.
  81. 81. Sellgren KL, Butala EJ, Gilmour BP, Randell SH, Grego S. A biomimetic multicellular model of the airways using primary human cells. Lab Chip. 2014;14[17]:3349–58. pmid:25000964
  82. 82. Benam KH, Villenave R, Lucchesi C, Varone A, Hubeau C, Lee HH, et al. Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro. Nat Methods. 2016;13[2]:151–7. pmid:26689262