Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Host Microbe ; 20(2): 250-8, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27512905

ABSTRACT

The blockade of phagolysosomal fusion is considered a critical mycobacterial strategy to survive in macrophages. However, viable mycobacteria have been observed in phagolysosomes during infection of cultured macrophages, and mycobacteria have the virulence determinant MarP, which confers acid resistance in vitro. Here we show in mice and zebrafish that innate macrophages overcome mycobacterial lysosomal avoidance strategies to rapidly deliver a substantial proportion of infecting bacteria to phagolysosomes. Exploiting the optical transparency of the zebrafish, we tracked the fates of individual mycobacteria delivered to phagosomes versus phagolysosomes and discovered that bacteria survive and grow in phagolysosomes, though growth is slower. MarP is required specifically for phagolysosomal survival, making it an important determinant for the establishment of mycobacterial infection in their hosts. Our work suggests that if pathogenic mycobacteria fail to prevent lysosomal trafficking, they tolerate the resulting acidic environment of the phagolysosome to establish infection.


Subject(s)
Anti-Bacterial Agents/metabolism , Carboxylic Acids/metabolism , Lysosomes/microbiology , Macrophages/microbiology , Microbial Viability/drug effects , Mycobacterium marinum/physiology , Stress, Physiological , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Host-Pathogen Interactions , Mice, Inbred C57BL , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/drug effects , Mycobacterium marinum/growth & development , Virulence Factors/genetics , Virulence Factors/metabolism , Zebrafish
2.
Nature ; 505(7482): 218-22, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24336213

ABSTRACT

The evolutionary survival of Mycobacterium tuberculosis, the cause of human tuberculosis, depends on its ability to invade the host, replicate, and transmit infection. At its initial peripheral infection site in the distal lung airways, M. tuberculosis infects macrophages, which transport it to deeper tissues. How mycobacteria survive in these broadly microbicidal cells is an important question. Here we show in mice and zebrafish that M. tuberculosis, and its close pathogenic relative Mycobacterium marinum, preferentially recruit and infect permissive macrophages while evading microbicidal ones. This immune evasion is accomplished by using cell-surface-associated phthiocerol dimycoceroserate (PDIM) lipids to mask underlying pathogen-associated molecular patterns (PAMPs). In the absence of PDIM, these PAMPs signal a Toll-like receptor (TLR)-dependent recruitment of macrophages that produce microbicidal reactive nitrogen species. Concordantly, the related phenolic glycolipids (PGLs) promote the recruitment of permissive macrophages through a host chemokine receptor 2 (CCR2)-mediated pathway. Thus, we have identified coordinated roles for PDIM, known to be essential for mycobacterial virulence, and PGL, which (along with CCR2) is known to be associated with human tuberculosis. Our findings also suggest an explanation for the longstanding observation that M. tuberculosis initiates infection in the relatively sterile environment of the lower respiratory tract, rather than in the upper respiratory tract, where resident microflora and inhaled environmental microbes may continually recruit microbicidal macrophages through TLR-dependent signalling.


Subject(s)
Immune Evasion , Macrophages/microbiology , Membrane Lipids/metabolism , Mycobacterium/physiology , Animals , Female , Glycolipids/immunology , Glycolipids/metabolism , Lipids/biosynthesis , Lipids/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mycobacterium/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/physiology , Receptors, CCR2/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Virulence/immunology , Zebrafish/microbiology
3.
Cell Rep ; 2(1): 175-84, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22840407

ABSTRACT

Treatment of tuberculosis, like other infectious diseases, is increasingly hindered by the emergence of drug resistance. Drug discovery efforts would be facilitated by facile screening tools that incorporate the complexities of human disease. Mycobacterium marinum-infected zebrafish larvae recapitulate key aspects of tuberculosis pathogenesis and drug treatment. Here, we develop a model for rapid in vivo drug screening using fluorescence-based methods for serial quantitative assessment of drug efficacy and toxicity. We provide proof-of-concept that both traditional bacterial-targeting antitubercular drugs and newly identified host-targeting drugs would be discovered through the use of this model. We demonstrate the model's utility for the identification of synergistic combinations of antibacterial drugs and demonstrate synergy between bacterial- and host-targeting compounds. Thus, the platform can be used to identify new antibacterial agents and entirely new classes of drugs that thwart infection by targeting host pathways. The methods developed here should be widely applicable to small-molecule screens for other infectious and noninfectious diseases.


Subject(s)
Antitubercular Agents/isolation & purification , Drug Discovery/methods , High-Throughput Screening Assays/methods , Animal Husbandry , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/isolation & purification , Antitubercular Agents/administration & dosage , Automation, Laboratory , Cryoanesthesia/methods , Cryoanesthesia/veterinary , Drug Discovery/instrumentation , Drug Synergism , Fluorometry/instrumentation , Fluorometry/methods , High-Throughput Screening Assays/instrumentation , Larva/growth & development , Larva/physiology , Models, Biological , Reproducibility of Results , Time Factors , Zebrafish/growth & development , Zebrafish/physiology
4.
Cell ; 145(1): 39-53, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21376383

ABSTRACT

Treatment of tuberculosis, a complex granulomatous disease, requires long-term multidrug therapy to overcome tolerance, an epigenetic drug resistance that is widely attributed to nonreplicating bacterial subpopulations. Here, we deploy Mycobacterium marinum-infected zebrafish larvae for in vivo characterization of antitubercular drug activity and tolerance. We describe the existence of multidrug-tolerant organisms that arise within days of infection, are enriched in the replicating intracellular population, and are amplified and disseminated by the tuberculous granuloma. Bacterial efflux pumps that are required for intracellular growth mediate this macrophage-induced tolerance. This tolerant population also develops when Mycobacterium tuberculosis infects cultured macrophages, suggesting that it contributes to the burden of drug tolerance in human tuberculosis. Efflux pump inhibitors like verapamil reduce this tolerance. Thus, the addition of this currently approved drug or more specific efflux pump inhibitors to standard antitubercular therapy should shorten the duration of curative treatment.


Subject(s)
Drug Tolerance , Macrophages/microbiology , Mycobacterium marinum/physiology , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Animals , Antitubercular Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Disease Models, Animal , Granuloma/physiopathology , Humans , Larva/microbiology , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/metabolism , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium Infections, Nontuberculous/physiopathology , Mycobacterium marinum/drug effects , Tuberculosis/drug therapy , Tuberculosis/pathology , Tuberculosis/physiopathology , Verapamil/pharmacology , Zebrafish/microbiology
5.
J Infect Dis ; 198(12): 1851-5, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18983252

ABSTRACT

Although tuberculous granulomas, which are composed of infected macrophages and other immune cells, have long been considered impermeable structures, recent studies have shown that superinfecting Mycobacterium marinum traffic rapidly to established fish and frog granulomas by host-mediated and Mycobacterium-directed mechanisms. The present study shows that superinfecting Mycobacterium tuberculosis and Mycobacterium bovis bacille Calmette-Guérin similarly home to established granulomas in mice. Furthermore, 2 prominent mycobacterial virulence determinants, Erp and ESX-1, do not affect this cellular trafficking. These findings suggest that homing of infected macrophages to sites of infection is a general feature of the pathogenesis of tuberculosis and has important consequences for therapeutic strategies.


Subject(s)
Bacterial Proteins/genetics , Granuloma/microbiology , Mycobacterium Infections/microbiology , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/physiology , Superinfection , Animals , Bacterial Proteins/metabolism , Chemotaxis , Female , Gene Expression Regulation, Bacterial , Mice , Mice, Inbred C57BL , Mycobacterium bovis/genetics , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Virulence
6.
Infect Immun ; 74(6): 3125-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16714540

ABSTRACT

The Mycobacterium tuberculosis exported repetitive protein (Erp) is a virulence determinant required for growth in cultured macrophages and in vivo. To better understand the role of Erp in Mycobacterium pathogenesis, we generated a mutation in the erp homologue of Mycobacterium marinum, a close genetic relative of M. tuberculosis. erp-deficient M. marinum was growth attenuated in cultured macrophage monolayers and during chronic granulomatous infection of leopard frogs, suggesting that Erp function is similarly required for the virulence of both M. tuberculosis and M. marinum. To pinpoint the step in infection at which Erp is required, we utilized a zebrafish embryo infection model that allows M. marinum infections to be visualized in real-time, comparing the erp-deficient strain to a DeltaRD1 mutant whose stage of attenuation was previously characterized in zebrafish embryos. A detailed microscopic examination of infected embryos revealed that bacteria lacking Erp were compromised very early in infection, failing to grow and/or survive upon phagocytosis by host macrophages. In contrast, DeltaRD1 mutant bacteria grow normally in macrophages but fail to induce host macrophage aggregation and subsequent cell-to-cell spread. Consistent with these in vivo findings, erp-deficient but not RD1-deficient bacteria exhibited permeability defects in vitro, which may be responsible for their specific failure to survive in host macrophages.


Subject(s)
Bacterial Proteins/physiology , Mycobacterium marinum/pathogenicity , Animals , Cell Wall , Cells, Cultured , Embryo, Mammalian/microbiology , Embryo, Nonmammalian , Male , Mice , Mycobacterium marinum/growth & development , Permeability , Rana pipiens , Virulence , Zebrafish
7.
Curr Protoc Microbiol ; Chapter 10: Unit 10B.2, 2006 Dec.
Article in English | MEDLINE | ID: mdl-18770575

ABSTRACT

Mycobacterium marinum infection of poikilothermic animals, such as fish and frogs, results in chronic granulomatous diseases that bear many similarities to mycobacterioses in mammals, including tuberculosis. This unit describes three animal models of M. marinum infection that can be used to study basic aspects of Mycobacterium-host interactions and granuloma development, as well as trafficking of immune cells in host tissues. Protocols are included that describe intraperitoneal infection of adult leopard frogs (Rana pipiens) and zebrafish (Danio rerio). Protocols also describe subsequent monitoring of the infection by enumeration of bacterial cfu, mean time to death, or visual examination of infected tissue using both conventional histological stains and fluorescence microscopy of fluorescently marked bacteria. Furthermore, protocols are included that describe the infection of embryonic zebrafish and the subsequent analysis of the infection in real time using DIC and fluorescence microscopy.


Subject(s)
Disease Models, Animal , Mycobacterium Infections, Nontuberculous/mortality , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium marinum/pathogenicity , Rana pipiens/microbiology , Zebrafish/microbiology , Animals , Embryo, Nonmammalian/microbiology , Host-Pathogen Interactions , Humans , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/isolation & purification , Zebrafish/embryology
8.
Nat Immunol ; 5(8): 828-35, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15220915

ABSTRACT

A central paradox of tuberculosis immunity is that reinfection and bacterial persistence occur despite vigorous host immune responses concentrated in granulomas, which are organized structures that form in response to infection. Prevailing models attribute reinfection and persistence to bacterial avoidance of host immunity via establishment of infection outside primary granulomas. Alternatively, persistence is attributed to a gradual bacterial adaptation to evolving host immune responses. We show here that superinfecting Mycobacterium marinum traffic rapidly into preexisting granulomas, including their caseous (necrotic) centers, through specific mycobacterium-directed and host cell-mediated processes, yet adapt quickly to persist long term therein. These findings demonstrate a failure of established granulomas, concentrated foci of activated macrophages and antigen-specific immune effector cells, to eradicate newly deposited mycobacteria not previously exposed to host responses.


Subject(s)
Granuloma/immunology , Mycobacterium Infections, Nontuberculous/immunology , Superinfection/immunology , Tuberculosis/immunology , Animals , Disease Models, Animal , Gene Products, gag/biosynthesis , Liver/immunology , Liver/microbiology , Liver/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Mycobacterium marinum/immunology , Rana pipiens , Salmonella Infections/immunology , Salmonella arizonae/immunology , Spleen/immunology , Spleen/microbiology , Spleen/pathology , Tuberculosis/pathology , Zebrafish
9.
Annu Rev Microbiol ; 57: 641-76, 2003.
Article in English | MEDLINE | ID: mdl-14527294

ABSTRACT

Pathogenic mycobacteria, including the causative agents of tuberculosis and leprosy, are responsible for considerable morbidity and mortality worldwide. A hallmark of these pathogens is their tendency to establish chronic infections that produce similar pathologies in a variety of hosts. During infection, mycobacteria reside in macrophages and induce the formation of granulomas, organized immune complexes of differentiated macrophages, lymphocytes, and other cells. This review summarizes our understanding of Mycobacterium-host cell interactions, the bacterial-granuloma interface, and mechanisms of bacterial virulence and persistence. In addition, we highlight current controversies and unanswered questions in these areas.


Subject(s)
Macrophages/microbiology , Mycobacterium Infections/microbiology , Mycobacterium/physiology , Animals , Disease Models, Animal , Genes, Bacterial , Granuloma/immunology , Granuloma/microbiology , Humans , Mycobacterium/pathogenicity , Mycobacterium avium Complex/pathogenicity , Mycobacterium avium Complex/physiology , Mycobacterium leprae/pathogenicity , Mycobacterium leprae/physiology , Mycobacterium marinum/pathogenicity , Mycobacterium marinum/physiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/physiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...