Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Critical ReviewsTM in Eukaryotic Gene Expression, 21(4):337-346 (2011)

New Insights into the Pathogenesis of Tuberculosis


Revealed by Mycobacterium marinum:
The Zebrafish Model from the Systems
Biology Perspective
Wanyan Denga, Xiemei Tangb, Manmei Houb, Chunmei Lib & Jianping Xiea,b,*

Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Enviroment and Bio-Resource
a,b

of the Three Gorges Area, School of Life Sciences, Southwest University, Beibei, Chongqing, China

*Address all correspondence to: Jianping Xie; Tel./Fax: 862368367108; Email: jianpingxie@vip.sina.com; jianpingxie@swu.edu.cn; georgex@swu.edu.cn

ABSTRACT: Tuberculosis remains a worldwide health concern, largely due to the emergence of multi-drug-
resistant (MDR) and extensive-drug-resistant (XDR) Mycobacterium tuberculosis co-infection with HIV. The
exact mechanism of Mycobacterium virulence, pathogenesis, and persistence is not fully understood. The hall-
mark of tuberculosis, granulomas are promoted by Mycobacterium virulence factors, and they have long
been considered a structural advantage to the host. However, this traditional view has been challenged re-
cently, largely due to the evidence originating from the M. marinum–zebrafish model. As a genetically trac-
table model, zebrafish provide unprecedented opportunities to address the pathogenesis of tuberculosis from a
systems biology perspective. The latest data from this model are summarized in this review, special attention is
given to the shared pathway and network between zebrafish and humans. This research serves to deepen our un-
derstanding of this complex process and to promote the discovery of better countermeasures against tuberculosis.

KEY WORDS: Tuberculosis, pathogenesis, M. marinum–zebrafish model, systems biology.

I. INTRODUCTION drawbacks of the poorly organized macrophage


aggregates that do not undergo necrosis seen in
Mycobacterium tuberculosis is the causative agent mouse models,2 the shortage of available immuno-
of tuberculosis (TB), a leading infectious disease. logical and molecular reagents seen in guinea pig
Approximately one-third of the global population and rabbit models,3–4 and the ethical and cost issues
is infected by this pathogen. Currently, the sole related to nonhuman primate models.3 The zebraf-
available vaccine and medications are problematic.1 ish model is more relevant to humans than that of
Novel measures to combat TB are urgently needed. amoeba (Dictyostelium discoideum) and fruit flies
Insight into the biology of M. tuberculosis is a pre- (Drosophila melanogaster) for the pathogenesis of
requisite for future rational interventions. The read- TB.4
ily manipulated M. marinum, highly similar to M. The typical feature of TB is the formation of
tuberculosis in sequence, is a unique and beneficial granuloma necroses, which are clusters of infect-
surrogate of M. tuberculosis that serves to accelerate ed macrophages surrounded by additional macro-
our research efforts. phages, neutrophils, and lymphocytes.5 As the main
Animal models are crucial to discovering the battlefield of confrontation between pathogen and
pathogenesis of Mycobacterium, and zebrafish pro- host, the traditional role of granulomas is being
vide an ideal model for this purpose. As the native challenged.6 Once regarded as beneficial to the host,
host of M. marinum, zebrafish can circumvent the recent data cast some doubt on this paradigm. These

1045-4403/11/$35.00 © 2011 Begell House, Inc. www.begellhouse.com 337


338 Deng et al.

new data are summarized here. III. NEW INSIGHTS INTO THE
FUNCTIONS OF GRANULOMAS
II. THE ADVANTAGES OF THE
M. MARINUM–ZEBRAFISH MODEL Granulomas, aggregates of immune cells including
macrophages and lymphocytes, are the hallmark of
The M. marinum–zebrafish model has been well TB.15–17 Granulomas have long been viewed as pro-
established as a useful model to address the tective structures that physically “wall off” persist-
pathogenesis of Mycobacterium infections. The ing bacteria to maintain the function of neighboring
relatively short generation time of 4 h of M. mari- tissue and to effectively prevent the replication and
num in contrast with the >20-h generation time of transmission of the pathogen.17 Granulomas have
M. tuberculosis is a great advantage.7 The natu- been viewed as an adaptive structure that forms af-
ral hosts of M. marinum are some poikilothermic ter the initiation of adaptive immunity.17 The bacteria
animals, including leopard frog (Rana pipiens), load increases rapidly following the first 2 weeks
goldfish (Carassius auratus), and zebrafish (Da- of infection, and the plateau coincides with the de-
nio rerio). Mycobacterium marinum are less like- velopment of adaptive immunity in animal models
ly to infect humans because the optimum growth of TB.13 Mature granulomas represent a stalemate
temperature of M. marinum is comparatively or equilibrium between host and pathogen, demon-
lower.8 The successful zebrafish developmental strated by the arrest of progressive infection and en-
model has been expanded for use as a model for capsulation of some live bacteria.18 However, recent
the analysis of host–pathogen interactions dur- studies show that the role of granulomas is stage de-
ing infectious disease.9 Zebrafish larvae are op- pendent. The early granulomas facilitate Mycobac-
tically transparent, allowing real-time monitor- terium transmission before the formation of adap-
ing of the early steps of M. marinum infection tive immunity,19 but they check the Mycobacterium
in live animals.10 The genome of M. marinum is growth during later stages.20 Recent studies prefer the
extremely conserved in this model, with an 85% view that granulomas serve as a place for bacterial
resemblance to human amino acid sequence; in growth and spread.7,21 First, granuloma formation co-
addition, the immune system of the zebrafish incides with accelerated bacterial expansion; it does
bears remarkable similarities to its mammalian not precede the expansion.22 Second, Mm lacking
counterparts.11 Zebrafish embryos have not yet the ESX-1/RD1 secretion system locus (DRD1 Mm)
developed an adaptive immune system, which is attenuated and is associated with poor granuloma
enables the investigation of the role of the innate formation.23 Finally, epithelioid granulomas develop
immune response during Mycobacterium disease within a few days of infection, prior to the presence
separately from the role of the adaptive system.12 of adaptive immunity.10 Real-time 3D-DIC (long-
Importantly, zebrafish produce caseating granu- term three-dimensional differential interference con-
lomas resembling human granulomas during M. trast) and fluorescence in vivo microscopy techniques
marinum infection.13 The apparent advantages of used at the whole-animal level have revealed that the
the M. marinum–zebrafish model over two other region of difference 1 (RD1) might be responsible
models (i.e., M. marinum–frog and M. marinum– for this subversive discovery.20 Mycobacterium RD1
goldfish) are the availability of genetic research can both enhance the recruitment of macrophages to
tools and real-time monitoring of the infection.10 the nascent granulomas and accelerate the pathogen-
The ever-increasing role of innate immunity in esis and transmission of Mycobacterium by invad-
vertebrates against infection can be better ad- ing the early granulomas.20 In fact, the RD1 region
dressed using the M. marinum–zebrafish model. A can initiate granuloma formation.19 ESAT-6 (protein
high-throughput screening system can be directly 6-kD early secreted antigenic target), encoded by the
executed in the M. marinum–zebrafish infection RD1 region, can induce matrix metalloproteinase-9
model, especially for screening compounds.14 (MMP9) in epithelial cells neighboring infected mac-

Critical ReviewsTM in Eukaryotic Gene Expression


New insight into Mycobacterium marinum- zebrafish model 339

rophages. MMP9 has been shown to enhance the re- M. marinum MMAR_2770 mutant is a biotin auxo-
cruitment of macrophages. The disruption of MMP9 troph and is defective in growth in macrophages
can attenuate granuloma formation and bacterial and zebrafish. In reconstructing MMAR_2770 or
growth. Therefore, interception of epithelial MMP9 its M. tuberculosis homolog Rv1882c (82% iden-
production might be promising for host-targeting TB tity in amino acid sequence) to the MMAR_2770
therapeutics. Whether this scenario exactly mirrors mutant, the phenotype was restored,25 suggesting
the M. tuberculosis infection in humans, however, that MMAR_2770 and its M. tuberculosis homo-
remains to be determined. log Rv1882c serve as new enzymes involved in
biotin biosynthesis and as potential targets for the
IV. NEW GENES AND FUNCTIONS development of antibiotics against Mycobacte-
INVOLVED IN MYCOBACTERIUM rium. An attenuated mutant TesA was found from
INFECTION a M. marinum transposon mutant library using a
Dictyostelium discoideum assay.26 TesA is required
Many well-characterized and new virulence fac- for the synthesis of phthioglycol diphthioceranates
tors crucial for Mycobacterium pathogenesis and phenolic glycolipids because both lipids were
have been discovered based on research using absent in TesA mutants. This mutant was found
the M. marinum–zebrafish model,6 such as those to be highly attenuated in zebrafish embryos, but
located in Esx-1 cluster EspA (MMAR_4166, virulence was retained when bacteria were injected
Rv3616c), EspB (MMAR_5457, Rv3881c), into the notochord,26 suggesting the high suscep-
EsxB (MMAR_0187, Rv3874), and ESAT-6 tibility of notochord to Mycobacterium infection.
(MMAR_0188, Rv3875); secreted repeat proteins
Erp (MMAR_5374, Rv3810); phagosome matura- B. Host Genes with New Functions
tion inhibiting protein PmiA (MMAR_3386); Mel1,
Mel2 gene locus protein MelA (MMAR_2864, Lta4h encode leukotriene A4 hydrolase, which
Rv2560), MelB (MMAR_2165, Rv2566), MelC catalyzes the final step in the synthesis of leukot-
(MMAR_2164, Rv2567), MelD (MMAR_2163, riene B4 (LTB4), a potent chemoattractant and
Rv2568c), MelE (MMAR_2162, Rv2569c), MelF proinflammatory eicosanoid. A zebrafish lta4h mu-
(MMAR_2864, Rv1936), MelG (MMAR_2865, tant larvae showed innate susceptibility to M. mari-
Rv1937), Mel (MMAR_2866, EphB (Rv1938)), num in a forward genetic screen. Lta4h mutations
MelI (MMAR_2867, Rv1939), MelJ (MMAR_2868, confer hypersusceptibility independent of LTB4
Rv1940), MelK (MMAR_2869, Rv1941); and reduction by redirecting eicosanoid substrates to
methadone acyl-b-ketoacyl-acyl carrier protein anti-inflammatory lipoxins A4 (LXA4).27 In addi-
synthase BKasB (MMAR_3339, Rv2246). In this tion, both TB and multibacillary leprosy protec-
section we highlight some specific findings from tion are associated with heterozygosity for LTA4H
both the host and the Mycobacterium. polymorphisms correlated with differential LTB4
production. Six LTA4H SNPs in 692 cases and 759
A. Genes with Enzymatic Activity controls followed the Hardy-Weinberg equilibrium
(HWE) in the control series, but significantly de-
Vitamins (i.e., vitamin B7 or H) often serve as viated from HWE in the case series. At each site
cofactor of enzymatic carboxylation reactions in- among the cases, fewer heterozygotes were present
volved in the carboxylation of fatty acid synthesis, than expected by the HWE prediction, suggesting
amino acid metabolism, and glycol-metabolism that heterozygosity at this locus might be protec-
necessary to all living organisms.24 Enzymes in- tive against TB.27 However, analyses of six LTA4H
volved in biotin biosynthesis are potential targets gene polymorphisms in samples collected from
for the development of antibiotics; biotin biosyn- Russia yielded contrasting results, suggesting that
thesis is unique to plants and microorganisms. The common polymorphisms in the LTA4H gene might

Volume 21, Number 4, 2011


340 Deng et al.

not play a major role in susceptibility to clinical of granulomas.13 The most well-studied genetic
pulmonary TB.28 The Russian sample is European locus involved in Mycobacterium virulence and
in origin and is distinct from the Vietnamese and granuloma formation is probably RD1, which in-
Nepali samples associated with TB and leprosy.27 cludes nine genes and is deleted in all attenuated
The exact roles of LTA4H SNPs in TB susceptibil- M. bovis bacille Calmette-Guérin (BCG) vaccine
ity remain to be validated in more samples from strains.39 The nine genes of the RD1 region are part
diverse populations. of the ESX-1 locus, which encodes a specialized
system responsible for the secretion of a number
C. Genes Affect the Transmission of proteins, including the important virulence fac-
of M. marinum tors ESAT-6 and CFP-10.40 It has been reported
that ESAT-6 is involved in the disruption of mem-
Defects in the espL, located in ESX-1 gene clus- branes; consequently, the protein has been asso-
ter, can reduce early granuloma formation.29 The ciated with the translocation of Mycobacterium
ESX-1 secretion system encoding genes include from the phagosome into the cytosol and subse-
ESAT-6, CFP-10, EspA, and EspB (Mh3881c).30–32 quent intercellular spread.33,41 Reintroduction of
ESAT-6 may cause membrane pore formation in an intact version of RD1 into M. bovis BCG re-
the MCV (Mycobacterium-containing vacuole), sulted in increased bacterial growth and formation
facilitating M. marinum escape from the vacuole of granuloma-like structures in immunodeficient
into the host-cell cytosol and to disseminate.33 The mice,42 whereas deletion of RD1 from M. tuber-
M. marinum locus of two genes (iipA and iipB) is culosis led to attenuation of both bacterial replica-
required for both invasion and intracellular sur- tion and granuloma formation in a mouse infection
vival within macrophages. Mycobacterium tuber- model.43 Mycobacterium marinum shows delayed
culosis Rv1477 and Rv1478 are homologs of the granuloma formation in zebrafish when RD1 is
iipA and iipB gene, Rv1477, fully complementing absent.13,22 The disruption of host MMP9 (Matrix
the Iip mutant for infectivity in vivo, as well as for metalloproteinase-9) attenuated granuloma forma-
invasion and intracellular persistence in macro- tion and bacterial growth. Thus, host MMP9 does
phages. Rv1478 only partially complemented the not inhibit but rather promotes the transmission of
Iip mutant in vivo and restored invasion but not bacterium and the progress of infection by affect-
intracellular growth in macrophages.34 While IipA ing the formation of early granulomas.19,20 Recent
and IipB differ at their N termini, they are highly research has shown that three new Mycobacteri-
similar throughout their C-terminal NLPC_p60 um factors (i.e., FAM53 (fadE33), FAM58 (espL
domains. The p60 domain of Rv1478 is fully func- (mmar_5456), espB(mmar_5457)) and FAM6
tional to replace that of Rv1477, suggesting that the 7(mmar_5425 and mmar_5426)) involved in the
N-terminal sequence of Rv1477 is required for full initial stages of granuloma are related to bacterial
virulence in vivo and in macrophages. Further mu- virulence.44 These new identified Mycobacterium
tations have demonstrated that both Arg-Gly-Asp factors may promote the formation of granulomas
(RGD) and Asp-Cys-Ser-Gly (DCSG) sequences structure, resulting in a reversal of the classical
in the p60 domain are required for function. view that granulomas are protective structures that
can defend against mycobacterial infection.
V. NEW MYCOBACTERIUM
EFFECTORS AND HOST TARGETS VI. SYSTEMS BIOLOGY INSIGHTS INTO THE
MYCOBACTERIUM INFECTION BASED ON
Host and high-risk environmental factors are cru- TRANSCRIPTOME
cial for the transmission of M. tuberculosiss.35 The
virulence factor may affect the dissemination of Digital gene expression profiling (DGE) combines
Mycobacterium36–38 by promoting the formation high-throughput sequencing with a high-perfor-

Critical ReviewsTM in Eukaryotic Gene Expression


New insight into Mycobacterium marinum- zebrafish model 341

FIGURE 1: Novel genes found to regulate metabolism and signaling crosslinking pathway.

mance computer analysis technique that can there- that encode enzymes involved in carbohydrate,
by detect the tissue-specific gene expression.45,46 alcohol, steroid, amino acid, and lipid metabo-
Using this technique, 159 genes were found to lism. They also included cytoskeletal protein
be robustly regulated by Mycobacterium infec- coding genes, tight junction proteins, solute car-
tion. Up-regulated genes include many known riers, and fatty acid binding proteins.47,48 These
components of inflammatory signaling, such as results suggest that deep-sequencing analysis
Mycobacterium-infection-related genes and differ- revealed the complexity of the host’s transcrip-
ent marker genes of the myeloid lineage that in ze- tional response to Mycobacterium infection.
brafish characterize response to infection.47 Novel Novel host genes regulated by Mycobacteri-
high-throughput, deep-sequencing technology has um were identified by comparing data from DGE
dramatically changed the way that the functional analysis with a previous multi-platform microar-
complexity of transcriptomes can be studied. My- ray (i.e., UniGene, RefSeq, and Ensembl). These
cobacterium-induced transcriptome changes in the genes include csf2rb (CSF2RB, colony stimulat-
zebrafish model were investigated using Solexa/Il- ing factor 2 receptor), cmklr1 (CMKLR1), sart3
lumina’s DGE system.48 (SART3, squamous cell carcinoma antigen rec-
Up-regulated zebrafish genes have been ognized by T cells 3), hspb8 (HspB8, heat shock
shown to involve immune-response-related tran- protein, alpha-crystallin-related, B8), LOC565410
scription factors, proinflammatory cytokines, (CAPG, capping protein), rhoae (RHOA),
and MHC class II proteins. Gene groups encod- zgc:171802 (GPR84, G protein-coupled recep-
ing proteolytic enzymes and lysosomal proton- tor 84), akap13 (AKAP13,A kinase anchor pro-
transporting ATPases were also enriched among tein 13), ltb4dh (LTB4DH,leukotriene B4 12-hy-
the up-regulated transcripts.47,48 Down-regulated droxydehydrogenase), LOC368614 (HLA-DPA1,
genes included genes involved in gene groups major histocompatibility complex, class II, DP

Volume 21, Number 4, 2011


342 Deng et al.

TABLE 3. The new identified functions of partial human homologs regulated by


Mycobacterium from DGE.

Zebrafish gene Human homology DGE Function Reference

Component of the MHC class II protein com-


LOC368614 HLA-DPA1 up [52]
plex involved in antigen presentation
Regulate cell proliferation and cytokine pro-
zgc: 112143 TNFAIP9 up [53]
duction
LTF may serve as a negative effector in-
zgc: 112154 LTF up [54]
volved in participate in NPC carcinogenesis
Functions as a chaperone in association with
hspb8 HspB8 down macroautophagy stimulator Bag3 in protein [55-56]
aggregation diseases
Contribute to negative regulation of immune
rhogb RHOG up responses Immunological function. Promote [49,57]
cell survival by the activation of PI3K and Akt
CFB associated with aHUS (atypical hemo-
LOC792364 CFB up [58]
lytic uremic syndrome)

alpha 1), LOC557797 (CD18, ITGB2-integrin, κB, ERK, AKt, JNKK, and Ca2+ pathway) (Figure
beta 2), zgc:86681, capg (CAPG, capping pro- 1). Another new regulated human homology that
tein), LOC563727 (MAP3K8, mitogen-activated may be associated with human genetic disorders
protein kinase kinase kinase 8), LOC794777 was modulated by Mycobacterium (Table 1). Im-
(TNFAIP2, tumor necrosis factor, alpha-induced portantly, few host genes identified from DGE di-
protein 2), tnip1 (TNIP1, TNFAIP3 interacting rectly related to TB caused by M. tuberculosis. For
protein 1), cul5 (CUL5, cullin 5), irgq2 (IRGM), example, human RhoG can activate both apoptotic
novel/rgl2 (RGL2, ral guanine nucleotide dissoci- and anti-apoptotic pathways through JNK and NF-
ation stimulator-like 2), LOC564859 (STK40, ser- kappaB-independent PI3K/Akt pathways, respec-
ine/threonine kinase 40), zgc:112143 (TNFAIP9, tively.49 Recent evidence has shown that the single
tumor necrosis factor alpha-induced protein 9), nucleotide polymorphism (SNPs) of the IRGM
il11a (IL11), LOC799527 (CXCR3), ptger2l (PT- promoter region (1208A/G) is associated with sus-
GER2, prostaglandin E receptor 2), tfa (LTF, lac- ceptibility to TB.50 In addition, IRGM has been
totransferrin), rhogb (RHOG, ras homolog gene shown to eliminate intracellular mycobacteria by
family, member G), LOC792364 (CFB, comple- inducing autophagy and reducing the intracellular
ment factor B), mxi1 (MXI1, MXI1-MAX interac- bacillary load.51 These results may suggest that
tor 1 isoform a), bcor (BCOR, BCL-6 interacting new molecules, identified by DGE based on the M.
corepressor isoform c), and LOC555512 (IFI44, marinum–zebrafish model, might have a function
interferon-induced protein 44). in immune defense and might facilitate the Myco-
Some new molecules may play important roles bacterium evasion of host immunity.
in the crosslinking response of three key metabo-
lism processes (i.e., glycolysis, protein synthesis VII. CONCLUSION
and degradation, and fatty acid oxidation) and in
the important signaling cascade pathways (i.e., NF- The M. marinum–zebrafish model provides tre-

Critical ReviewsTM in Eukaryotic Gene Expression


New insight into Mycobacterium marinum- zebrafish model 343

mendously fresh vision for the development of mechanism from the zebra fish-Mycobacterium mari-
forthcoming therapies for TB. This model may num model—a review]. Wei Sheng Wu Xue Bao.
2010;50(1):15–22.
have transformed our traditional understanding of
9. Phelps HA, Neely MN, Evolution of the zebrafish model:
the roles of granulomas. With it, multiple candidate from development to immunity and infectious disease.
genes can be scrutinized. This model has helped to Zebrafish. 2005;2(2):87–103.
identify the virulence factor with unknown func- 10. Davis JM, Clay H, Lewis JL, Ghori N, Herbomel P,
tion. Although seemingly independent, these pro- Ramakrishnan L. Real-time visualization of mycobac-
teins may interplay during the pathogenesis of My- terium-macrophage interactions leading to initiation of
granuloma formation in zebrafish embryos. Immunity.
cobacterium. The M. marinum–zebrafish model,
2002;17(6):693–702.
together with other high-throughput and high-con- 11. Meeker ND, Trede NS. Immunology and zebrafish:
tent screening methodologies, are sure to expedite spawning new models of human disease. Dev Comp Im-
the discovery of novel antibiotics against TB. munol. 2008;32(7):745–57.
12. Langenau DM, Ferrando AA, Traver D, Kutok JL, He-
ACKNOWLEDGEMENT zel JP, Kanki JP, Zon LI, Look AT, Trede NS. In vivo
tracking of T cell development, ablation, and engraft-
ment in transgenic zebrafish. Proc Natl Acad Sci U S A.
This work was supported by the National Mega-
2004;101(19):7369–74.
project for Key Infectious Disease (Grant No. 13. Swaim LE, Connolly LE, Volkman HE, Humbert O,
2012ZX10003-003), the Fundamental Research Born DE, Ramakrishnan L. Mycobacterium marinum
Funds for the Central Universities (Grant No. XD- infection of adult zebrafish causes caseating granuloma-
JK2009A003), the National Natural Science Foun- tous tuberculosis and is moderated by adaptive immu-
dation (Grant No. 81071316), and the Excellent nity. Infect Immun. 2006;74(11):6108–17.
14. Carvalho R, de Sonneville J, Stockhammer OW, Savage
PhD Thesis Fellowship of Southwest University
NDL, Veneman WJ, Ottenhoff THM, Dirks RP, Meijer
(Grant Nos. kb2009010 and ky2009009). AH, Spaink HP. A high-throughput screen for tuberculo-
sis progression. PLoS One. 2011;6(2):e16779.
REFERENCES 15. Adams DO. The granulomatous inflammatory response.
A review. Am J Pathol. 1976;84(1):164–92.
1. Russell DG, Barry CE 3rd, Flynn JL. Tuberculosis: 16. Egen JG, Rothfuchs AG, Feng CG, Winter N, Sher A,
what we don’t know can, and does, hurt us. Science. Germain RN. Macrophage and T cell dynamics during
2010;328(5980):852–6. the development and disintegration of mycobacterial
2. Pozos TC, Ramakrishnan L. New models for the study of granulomas. Immunity. 2008;28(2):271–84.
Mycobact-erium–host interactions. Curr Opin Immunol. 17. Saunders BM, Cooper AM. Restraining mycobacteria:
2004;16(4):499–505. role of granulomas in mycobacterial infections. Immunol
3. Capuano SV 3rd, Croix DA, Pawar S, Zinovik A, My- Cell Biol. 2000;78(4):334–41.
ers A, Lin PL, Bissel S, Fuhrman C, Klein E, Flynn JL. 18. Bold TD, Ernst JD. Who benefits from granulomas, my-
Experimental Mycobacterium tuberculosis infection of cobacteria or host? Cell. 2009;136(1):17–9.
cynomolgus macaques closely resembles the various 19. Volkman HE, Pozos TC, Zheng J, Davis JM, Rawls JF,
manifestations of human M. tuberculosis infection. Infect Ramakrishnan L. Tuberculous granuloma induction via
Immun. 2003;71(10):5831–44. interaction of a bacterial secreted protein with host epi-
4. Dionne MS, Ghori N, Schneider DS. Drosophila mela- thelium. Science. 2010;327(5964):466–9.
nogaster is a genetically tractable model host for Myco- 20. Davis JM, Ramakrishnan L. The role of the granuloma in
bacterium marinum. Infect Immun. 2003;71(6):3540–50. expansion and dissemination of early tuberculous infec-
5. Russell DG. Who puts the tubercle in tuberculosis? Nat tion. Cell. 2009;136(1):37–49.
Rev Microbiol. 2007;5(1):39–47. 21. Lesley R, Ramakrishnan L. Insights into early mycobac-
6. Meijer AH, Spaink HP. Host-pathogen interactions made terial pathogenesis from the zebrafish. Curr Opin Micro-
transparent with the zebrafish model. Curr Drug Targets. biol. 2008;11(3):277–83.
2011;12(7):1000–17. 22. Volkman HE, Clay H, Beery D, Chang JC, Sherman DR,
7. Tobin DM, Ramakrishnan L. Comparative pathogenesis Ramakrishnan L. Tuberculous granuloma formation is
of Mycobacterium marinum and Mycobacterium tuber- enhanced by a mycobacterium virulence determinant.
culosis. Cell Microbiol. 2008;10(5):1027–39. PLoS Biol. 2004;2(11):e367.
8. Hou M, Xie J. [Insight into tuberculosis pathogenic 23. DiGiuseppe Champion, PA, Cox JS. Protein se-

Volume 21, Number 4, 2011


344 Deng et al.

cretion systems in mycobacteria. Cell Microbiol. sion and intracellular persistence in macrophages. Infect
2007;9(6):1376–84. Immun. 2006;74(3):1757–67.
24. Knowles JR. The mechanism of biotin-dependent en- 35. Nava-Aguilera E, Andersson N, Harris E, Mitchell S,
zymes. Annu Rev Biochem. 1989;58:195–221. Hamel C, Shea B, López-Vidal Y, Villegas-Arrizón A,
25. Yu J, Niu C, Wang D, Li M, Teo W, Sun G, Wang J, Liu J, Morales-Pérez A. Risk factors associated with recent
Gao Q. MMAR_2770, a new enzyme involved in biotin transmission of tuberculosis: systematic review and
biosynthesis, is essential for the growth of Mycobacte- meta-analysis. Int J Tuberc Lung Dis. 2009;13(1):17–26.
rium marinum in macrophages and zebrafish. Microbes 36. Valway SE, Sanchez MP, Shinnick TF, Orme I, Agerton
Infect. 2011;13(1):33–41. T, Hoy D, Jones JS, Westmoreland H, Onorato IM. An
26. Alibaud L, Rombouts Y, Trivelli X, Burguière A, Cirillo outbreak involving extensive transmission of a virulent
SL, Cirillo JD, Dubremetz JF, Guérardel Y, Lutfalla G, strain of Mycobacterium tuberculosis. N Engl J Med.
Kremer L. A Mycobacterium marinum TesA mutant de- 1998;338(10):633–9.
fective for major cell wall-associated lipids is highly at- 37. Nicol MP, Wilkinson RJ, The clinical consequences of
tenuated in Dictyostelium discoideum and zebrafish em- strain diversity in Mycobacterium tuberculosis. Trans R
bryos. Mol Microbiol. 2011. Soc Trop Med Hyg. 2008;102(10):955–65.
27. Tobin DM, Vary JC Jr, Ray JP, Walsh GS, Dunstan SJ, 38. van der Spuy GD, Kremer K, Ndabambi SL, Beyers
Bang ND, Hagge DA, Khadge S, King MC, Hawn TR, N, Dunbar R, Marais BJ, van Helden PD, Warren RM.
Moens CB, Ramakrishnan L. The lta4h locus modulates Changing Mycobacterium tuberculosis population high-
susceptibility to mycobacterial infection in zebrafish and lights clade-specific pathogenic characteristics. Tubercu-
humans. Cell. 2010;140(5):717–30. losis (Edinb). 2009;89(2):120–5.
28. Curtis J, Kopanitsa L, Stebbings E, Speirs A, Ignatyeva 39. Mahairas GG, Sabo PJ, Hickey MJ, Singh DC, Stover
O, Balabanova Y, Nikolayevskyy V, Hoffner S, Horst- CK. Molecular analysis of genetic differences between
mann R, Drobniewski F, Nejentsev S. Association analy- Mycobacterium bovis BCG and virulent M. bovis. J Bac-
sis of the LTA4H gene polymorphisms and pulmonary teriol. 1996;178(5):1274–82.
tuberculosis in 9115 subjects. Tuberculosis (Edinb). 40. Samten B, Wang X, Barnes PF. Mycobacterium tuber-
2011;91(1):22–5. culosis ESX-1 system-secreted protein ESAT-6 but not
29. Stoop EJ, Schipper T, Huber SK, Nezhinsky AE, Verbeek CFP10 inhibits human T-cell immune responses. Tuber-
FJ, Gurcha SS, Besra GS, Vandenbroucke-Grauls CM, culosis (Edinb). 2009;89 Suppl 1:S74–6.
Bitter W, van der Sar AM. Zebrafish embryo screen for 41. Gao LY, Guo S, McLaughlin B, Morisaki H, Engel JN,
mycobacterial genes involved in the initiation of granu- Brown EJ. A mycobacterial virulence gene cluster ex-
loma formation reveals a newly identified ESX-1 compo- tending RD1 is required for cytolysis, bacterial spreading
nent. Dis Model Mech. 2011. and ESAT-6 secretion. Mol Microbiol. 2004;53(6):1677–
30. Xu J, Laine O, Masciocchi M, Manoranjan J, Smith J, 93.
Du SJ, Edwards N, Zhu X, Fenselau C, Gao LY. A unique 42. Pym AS, Brodin P, Brosch R, Huerre M, Cole ST. Loss of
Mycobacterium ESX-1 protein co-secretes with CFP-10/ RD1 contributed to the attenuation of the live tuberculo-
ESAT-6 and is necessary for inhibiting phagosome matu- sis vaccines Mycobacterium bovis BCG and Mycobacte-
ration. Mol Microbiol. 2007;66(3):787–800. rium microti. Mol Microbiol. 2002;46(3):709–17.
31. McLaughlin B, Chon JS, MacGurn JA, Carlsson F, 43. Lewis KN, Liao R, Guinn KM, Hickey MJ, Smith S,
Cheng TL, Cox JS, Brown EJ. A mycobacterium ESX- Behr MA, Sherman DR. Deletion of RD1 from Myco-
1-secreted virulence factor with unique requirements for bacterium tuberculosis mimics bacille Calmette-Guerin
export. PLoS Pathog. 2007;3(8):e105. attenuation. J Infect Dis. 2003;187(1):117–23.
32. Fortune SM, Jaeger A, Sarracino DA, Chase MR, Sas- 44. Stoop EJ, Schipper T, Huber SK, Nezhinsky AE, Verbeek
setti CM, Sherman DR, Bloom BR, Rubin EJ. Mu- FJ, Gurcha SS, Besra GS, Vandenbroucke-Grauls CM,
tually dependent secretion of proteins required for Bitter W, van der Sar AM. Zebrafish embryo screen for
mycobacterial virulence. Proc Natl Acad Sci U S A. mycobacterial genes involved in the initiation of granu-
2005;102(30):10676–81. loma formation reveals a newly identified ESX-1 compo-
33. Smith J, Manoranjan J, Pan M, Bohsali A, Xu J, Liu J, nent. Dis Model Mech. 2011;4(4):526–36.
McDonald KL, Szyk A, LaRonde-LeBlanc N, Gao LY. 45. Qin YF, Fang HM, Tian QN, Bao ZX, Lu P, Zhao JM,
Evidence for pore formation in host cell membranes Mai J, Zhu ZY, Shu LL, Zhao L, Chen SJ, Liang F, Zhang
by ESX-1-secreted ESAT-6 and its role in Mycobacte- YZ, Zhang ST. Transcriptome profiling and digital gene
rium marinum escape from the vacuole. Infect Immun. expression by deep-sequencing in normal/regenerative
2008;76(12):5478–87. tissues of planarian Dugesia japonica. Genomics. 2011.
34. Gao LY, Pak M, Kish R, Kajihara K, Brown EJ. A myco- 46. Wang Z, Dong D, Ru B, Young RL, Han N, Guo T, Zhang
bacterial operon essential for virulence in vivo and inva- S. Digital gene expression tag profiling of bat digits pro-

Critical ReviewsTM in Eukaryotic Gene Expression


New insight into Mycobacterium marinum- zebrafish model 345

vides robust candidates contributing to wing formation. Umeda N, Kanamori A, Ochiai N, Miyazawa K, Sugi-
BMC Genomics. 2010;11:619. hara M, Hayashi T, Goto D, Ito S, Sumida T. Six-trans-
47. Meijer AH, Verbeek FJ, Salas-Vidal E, Corredor- membrane epithelial antigen of prostate4 (STEAP4) is
Adámez M, Bussman J, van der Sar AM, Otto GW, a tumor necrosis factor alpha-induced protein that regu-
Geisler R, Spaink HP. Transcriptome profiling of adult lates IL-6, IL-8, and cell proliferation in synovium from
zebrafish at the late stage of chronic tuberculosis due patients with rheumatoid arthritis. Mod Rheumatol. 2011
to Mycobacterium marinum infection. Mol Immunol. Jun 3 [Epub ahead of print].
2005;42(10):1185–203. 54. Zhang H, Feng X, Liu W, Jiang X, Shan W, Huang C,
48. Hegedus Z, Zakrzewska A, Agoston VC, Ordas A, Rácz Yi H, Zhu B, Zhou W, Wang L, Liu C, Zhang L, Jia W,
P, Mink M, Spaink HP, Meijer AH. Deep sequencing of Huang W, Li G, Shi J, Wanggou S, Yao K, Ren C. Under-
the zebrafish transcriptome response to mycobacterium lying mechanisms for LTF inactivation and its functional
infection. Mol Immunol. 2009;46(15):2918–30. analysis in nasopharyngeal carcinoma cell lines. J Cell
49. Murga C, Zohar M, Teramoto H, Gutkind JS. Rac1 and Biochem. 2011;112(7):1832–43.
RhoG promote cell survival by the activation of PI3K 55. Seidel K, Vinet J, den Dunnen WF, Brunt ER, Meister
and Akt, independently of their ability to stimulate JNK M, Boncoraglio A, Zijlstra MP, Boddeke HW, Rüb U,
and NF-kappaB. Oncogene. 2002;21(2):207–16. Kampinga HH, Carra S. The HSPB8-BAG3 chaperone
50. Che N, Li S, Gao T, Zhang Z, Han Y, Zhang X, Sun Y, complex is upregulated in astrocytes in the human brain
Liu Y, Sun Z, Zhang J, Ren W, Tian M, Li Y, Li W, Cheng affected by protein aggregation diseases. Neuropathol
J, Li C. Identification of a novel IRGM promoter single Appl Neurobiol. 2011 Jun 23 [Epub ahead of print].
nucleotide polymorphism associated with tuberculosis. 56. Carra S, Seguin SJ, Landry J. HspB8 and Bag3: a new
Clin Chim Acta. 2010;411(21–22):1645–9. chaperone complex targeting misfolded proteins to mac-
51. SB Singh, AS Davis, GA Taylor, V Deretic. Human roautophagy. Autophagy. 2008;4(2):237–9.
IRGM induces autophagy to eliminate intracellular my- 57. Vigorito E, Bell S, Hebeis BJ, Reynolds H, McAdam
cobacteria. Science. 2006;313(5792):1438–41. S, Emson PC, McKenzie A, Turner M. Immunological
52. Colledge L, Bennett CL, Reay PA, Blackburn CC. Rapid function in mice lacking the Rac-related GTPase RhoG.
constitutive generation of a specific peptide-MHC class II Mol Cell Biol. 2004;24(2):719–29.
complex from intact exogenous protein in immature mu- 58. Tawadrous H, Maga T, Sharma J, Kupferman J, Smith
rine dendritic cells. Eur J Immunol. 2002;32(11):3246– RJ, Schoeneman M. A novel mutation in the complement
55. factor B gene (CFB) and atypical hemolytic uremic syn-
53. Tanaka Y, Matsumoto I, Iwanami K, Inoue A, Minami R, drome. Pediatr Nephrol. 2010;25(5):947–51.

Volume 21, Number 4, 2011

You might also like