Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 39(3): 110715, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35443177

RESUMO

Tissue-resident innate lymphoid cells (ILCs) regulate tissue homeostasis, protect against pathogens at mucosal surfaces, and are key players at the interface of innate and adaptive immunity. How ILCs adapt their phenotype and function to environmental cues within tissues remains to be fully understood. Here, we show that Mycobacterium tuberculosis (Mtb) infection alters the phenotype and function of lung IL-18Rα+ ILC toward a protective interferon-γ-producing ILC1-like population. This differentiation is controlled by type 1 cytokines and is associated with a glycolytic program. Moreover, a BCG-driven type I milieu enhances the early generation of ILC1-like cells during secondary challenge with Mtb. Collectively, our data reveal how tissue-resident ILCs adapt to type 1 inflammation toward a pathogen-tailored immune response.


Assuntos
Imunidade Inata , Tuberculose , Citocinas , Humanos , Inflamação , Linfócitos
2.
Vaccine ; 38(6): 1416-1423, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31862194

RESUMO

Tuberculosis still claims more lives than any other pathogen, and a vaccine better than BCG is urgently needed. One of the challenges for novel TB vaccines is to protect against all Mycobacterium tuberculosis lineages, including the most virulent ones, such as the Beijing lineage. Here we developed a live attenuated M. tuberculosis mutant derived from GC1237, a Beijing strain responsible for tuberculosis outbreaks in the Canary Islands. The mutant strain is inactivated both in the Rv1503c gene, responsible for surface glycolipid synthesis, and in the two-component global regulator PhoPR. This double mutant is as safe as BCG in immunodeficient SCID mice. In immune-competent mice and guinea pigs, the mutant is as protective as BCG against M. tuberculosis strains of common lineage 4 (Euro-American). By contrast, in mice the vaccine is protective against a M. tuberculosis strain of lineage 2 (East-Asian, Beijing), while BCG is not. These results highlight differences in protection efficacy of live attenuated M. tuberculosis-derived vaccine candidates depending on their genetic background, and provide insights for the development of novel live vaccines against TB, especially in East-Asian countries where M. tuberculosis strains of the Beijing family are highly dominant.


Assuntos
Vacinas contra a Tuberculose/imunologia , Tuberculose , Animais , Vacina BCG , Cobaias , Camundongos , Camundongos SCID , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Tuberculose/prevenção & controle , Vacinas Atenuadas/imunologia
3.
Front Immunol ; 9: 2656, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30487801

RESUMO

Tuberculosis (TB), caused by the airborne bacterial pathogen Mycobacterium tuberculosis, remains a major source of morbidity and mortality worldwide. So far, the study of host-pathogen interactions in TB has mostly focused on the physiology and virulence of the pathogen, as well as, on the various innate and adaptive immune compartments of the host. Microbial organisms endogenous to our body, the so-called microbiota, interact not only with invading pathogens, but also with our immune system. Yet, the impact of the microbiota on host defense against M. tuberculosis remains poorly understood. In order to address this question, we adapted a robust and reproducible mouse model of microbial dysbiosis based on a combination of wide-spectrum antibiotics. We found that microbiota dysbiosis resulted in an increased early colonization of the lungs by M. tuberculosis during the first week of infection, correlating with an altered diversity of the gut microbiota during this time period. At the cellular level, no significant difference in the recruitment of conventional myeloid cells, including macrophages, dendritic cells and neutrophils, to the lungs could be detected during the first week of infection between microbiota-competent and -deficient mice. At the molecular level, microbiota depletion did not impact the global production of pro-inflammatory cytokines, such as interferon (IFN)γ, tumor necrosis factor (TNF)α and interleukin (IL)-1ß in the lungs. Strikingly, a reduced number of mucosal-associated invariant T (MAIT) cells, a population of innate-like lymphocytes whose development is known to depend on the host microbiota, was observed in the lungs of the antibiotics-treated animals after 1week of infection. These cells produced less IL-17A in antibiotics-treated mice. Notably, dysbiosis correction through the inoculation of a complex microbiota in antibiotics-treated animals reversed these phenotypes and improved the ability of MAIT cells to proliferate. Altogether, our results demonstrate that the host microbiota contributes to early protection of lung colonization by M. tuberculosis, possibly through sustaining the function(s) of MAIT cells. Our study calls for a better understanding of the impact of the microbiota on host-pathogen interactions in TB. Ultimately, this study may help to develop novel therapeutic approaches based on the use of beneficial microbes, or components thereof, to boost anti-mycobacterial immunity.


Assuntos
Pulmão , Microbiota/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar , Animais , Citocinas/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/patologia , Feminino , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Células T Invariantes Associadas à Mucosa/patologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Tuberculose Pulmonar/prevenção & controle
4.
PLoS Pathog ; 13(11): e1006752, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29176894

RESUMO

The unique ability of the tuberculosis (TB) bacillus, Mycobacterium tuberculosis, to persist for long periods of time in lung hypoxic lesions chiefly contributes to the global burden of latent TB. We and others previously reported that the M. tuberculosis ancestor underwent massive episodes of horizontal gene transfer (HGT), mostly from environmental species. Here, we sought to explore whether such ancient HGT played a part in M. tuberculosis evolution towards pathogenicity. We were interested by a HGT-acquired M. tuberculosis-specific gene set, namely moaA1-D1, which is involved in the biosynthesis of the molybdenum cofactor. Horizontal acquisition of this gene set was striking because homologues of these moa genes are present all across the Mycobacterium genus, including in M. tuberculosis. Here, we discovered that, unlike their paralogues, the moaA1-D1 genes are strongly induced under hypoxia. In vitro, a M. tuberculosis moaA1-D1-null mutant has an impaired ability to respire nitrate, to enter dormancy and to survive in oxygen-limiting conditions. Conversely, heterologous expression of moaA1-D1 in the phylogenetically closest non-TB mycobacterium, Mycobacterium kansasii, which lacks these genes, improves its capacity to respire nitrate and grants it with a marked ability to survive oxygen depletion. In vivo, the M. tuberculosis moaA1-D1-null mutant shows impaired survival in hypoxic granulomas in C3HeB/FeJ mice, but not in normoxic lesions in C57BL/6 animals. Collectively, our results identify a novel pathway required for M. tuberculosis resistance to host-imposed stress, namely hypoxia, and provide evidence that ancient HGT bolstered M. tuberculosis evolution from an environmental species towards a pervasive human-adapted pathogen.


Assuntos
Coenzimas/biossíntese , Transferência Genética Horizontal , Metaloproteínas/biossíntese , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Oxigênio/metabolismo , Tuberculose/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Hipóxia/metabolismo , Hipóxia/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Cofatores de Molibdênio , Mycobacterium/genética , Mycobacterium/metabolismo , Nitratos/metabolismo , Pteridinas , Tuberculose/metabolismo
5.
PLoS Pathog ; 10(2): e1003928, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586151

RESUMO

Mycobacterium tuberculosis is an intracellular pathogen. Within macrophages, M. tuberculosis thrives in a specialized membrane-bound vacuole, the phagosome, whose pH is slightly acidic, and where access to nutrients is limited. Understanding how the bacillus extracts and incorporates nutrients from its host may help develop novel strategies to combat tuberculosis. Here we show that M. tuberculosis employs the asparagine transporter AnsP2 and the secreted asparaginase AnsA to assimilate nitrogen and resist acid stress through asparagine hydrolysis and ammonia release. While the role of AnsP2 is partially spared by yet to be identified transporter(s), that of AnsA is crucial in both phagosome acidification arrest and intracellular replication, as an M. tuberculosis mutant lacking this asparaginase is ultimately attenuated in macrophages and in mice. Our study provides yet another example of the intimate link between physiology and virulence in the tubercle bacillus, and identifies a novel pathway to be targeted for therapeutic purposes.


Assuntos
Asparagina/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/metabolismo , Nitrogênio/metabolismo , Fagossomos/metabolismo , Estresse Fisiológico , Tuberculose/metabolismo , Animais , Cromatografia Líquida , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Técnicas de Inativação de Genes , Immunoblotting , Espectrometria de Massas , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Imunoeletrônica , Fagossomos/microbiologia
6.
Nat Chem Biol ; 9(11): 674-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24077180

RESUMO

Here we identify the amino acid transporter AnsP1 as the unique aspartate importer in the human pathogen Mycobacterium tuberculosis. Metabolomic analysis of a mutant with an inactive AnsP1 revealed that the transporter is essential for M. tuberculosis to assimilate nitrogen from aspartate. Virulence of the AnsP1 mutant is impaired in vivo, revealing that aspartate is a primary nitrogen source required for host colonization by the tuberculosis bacillus.


Assuntos
Ácido Aspártico/metabolismo , Mycobacterium tuberculosis/metabolismo , Nitrogênio/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo
7.
PLoS One ; 7(6): e38367, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22675550

RESUMO

The W-Beijing family of Mycobacterium tuberculosis (Mtb) strains is known for its high-prevalence and -virulence, as well as for its genetic diversity, as recently reported by our laboratories and others. However, little is known about how the immune system responds to these strains. To explore this issue, here we used reverse engineering and genome-wide expression profiling of human macrophage-like THP-1 cells infected by different Mtb strains of the W-Beijing family, as well as by the reference laboratory strain H37Rv. Detailed data mining revealed that host cell transcriptome responses to H37Rv and to different strains of the W-Beijing family are similar and overwhelmingly induced during Mtb infections, collectively typifying a robust gene expression signature ("THP1r2Mtb-induced signature"). Analysis of the putative transcription factor binding sites in promoter regions of genes in this signature identified several key regulators, namely STATs, IRF-1, IRF-7, and Oct-1, commonly involved in interferon-related immune responses. The THP1r2Mtb-induced signature appeared to be highly relevant to the interferon-inducible signature recently reported in active pulmonary tuberculosis patients, as revealed by cross-signature and cross-module comparisons. Further analysis of the publicly available transcriptome data from human patients showed that the signature appears to be relevant to active pulmonary tuberculosis patients and their clinical therapy, and be tuberculosis specific. Thus, our results provide an additional layer of information at the transcriptome level on mechanisms involved in host macrophage response to Mtb, which may also implicate the robustness of the cellular defense system that can effectively fight against genetic heterogeneity in this pathogen.


Assuntos
Perfilação da Expressão Gênica , Interferons/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Tuberculose/genética , Tuberculose/imunologia , Linhagem Celular , Humanos , Inflamação/genética , Inflamação/patologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Especificidade da Espécie , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transcriptoma/genética , Tuberculose/diagnóstico , Tuberculose/microbiologia
8.
Cell Host Microbe ; 10(3): 248-59, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21925112

RESUMO

Mycobacterium tuberculosis thrives within macrophages by residing in phagosomes and preventing them from maturing and fusing with lysosomes. A parallel transcriptional survey of intracellular mycobacteria and their host macrophages revealed signatures of heavy metal poisoning. In particular, mycobacterial genes encoding heavy metal efflux P-type ATPases CtpC, CtpG, and CtpV, and host cell metallothioneins and zinc exporter ZnT1, were induced during infection. Consistent with this pattern of gene modulation, we observed a burst of free zinc inside macrophages, and intraphagosomal zinc accumulation within a few hours postinfection. Zinc exposure led to rapid CtpC induction, and ctpC deficiency caused zinc retention within the mycobacterial cytoplasm, leading to impaired intracellular growth of the bacilli. Thus, the use of P(1)-type ATPases represents a M. tuberculosis strategy to neutralize the toxic effects of zinc in macrophages. We propose that heavy metal toxicity and its counteraction might represent yet another chapter in the host-microbe arms race.


Assuntos
ATPases Bacterianas Próton-Translocadoras/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/enzimologia , Tuberculose/metabolismo , Zinco/metabolismo , Animais , ATPases Bacterianas Próton-Translocadoras/genética , Células Cultivadas , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Tuberculose/microbiologia , Zinco/toxicidade
9.
PLoS Pathog ; 6(9): e1001100, 2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20844580

RESUMO

The ability of the tubercle bacillus to arrest phagosome maturation is considered one major mechanism that allows its survival within host macrophages. To identify mycobacterial genes involved in this process, we developed a high throughput phenotypic cell-based assay enabling individual sub-cellular analysis of over 11,000 Mycobacterium tuberculosis mutants. This very stringent assay makes use of fluorescent staining for intracellular acidic compartments, and automated confocal microscopy to quantitatively determine the intracellular localization of M. tuberculosis. We characterised the ten mutants that traffic most frequently into acidified compartments early after phagocytosis, suggesting that they had lost their ability to arrest phagosomal maturation. Molecular analysis of these mutants revealed mainly disruptions in genes involved in cell envelope biogenesis (fadD28), the ESX-1 secretion system (espL/Rv3880), molybdopterin biosynthesis (moaC1 and moaD1), as well as in genes from a novel locus, Rv1503c-Rv1506c. Most interestingly, the mutants in Rv1503c and Rv1506c were perturbed in the biosynthesis of acyltrehalose-containing glycolipids. Our results suggest that such glycolipids indeed play a critical role in the early intracellular fate of the tubercle bacillus. The unbiased approach developed here can be easily adapted for functional genomics study of intracellular pathogens, together with focused discovery of new anti-microbials.


Assuntos
Glicolipídeos/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/metabolismo , Fagossomos/fisiologia , Tuberculose/metabolismo , Tuberculose/patologia , Animais , Feminino , Macrófagos/citologia , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagocitose , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tuberculose/microbiologia
10.
Microbiology (Reading) ; 156(Pt 6): 1619-1631, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20185505

RESUMO

Eukaryotic-like Ser/Thr protein kinases (STPKs) are present in many bacterial species, where they control various physiological and virulence processes by enabling microbial adaptation to specific environmental signals. PknJ is the only member of the 11 STPKs identified in Mycobacterium tuberculosis that still awaits characterization. Here we report that PknJ is a functional kinase that forms dimers in vitro, and contains a single transmembrane domain. Using a high-density peptide-chip-based technology, multiple potential mycobacterial targets were identified for PknJ. We confirmed PknJ-dependent phosphorylation of four of these targets: PknJ itself, which autophosphorylates at Thr(168), Thr(171) and Thr(173) residues; the transcriptional regulator EmbR; the methyltransferase MmaA4/Hma involved in mycolic acid biosynthesis; and the dipeptidase PepE, whose encoding gene is located next to pknJ in the mycobacterial genome. Our results provide a number of candidate phospho-targets for PknJ and possibly other mycobacterial STPKs that could be studied to investigate the role of STPKs in M. tuberculosis physiology and virulence.


Assuntos
Mycobacterium tuberculosis/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Dimerização , Camundongos , Dados de Sequência Molecular , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Alinhamento de Sequência , Serina/metabolismo , Transdução de Sinais , Treonina/metabolismo , Tuberculose/microbiologia , Virulência
11.
J Exp Med ; 206(10): 2205-20, 2009 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-19770268

RESUMO

The C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) mediates the innate immune recognition of microbial carbohydrates. We investigated the function of this molecule in the host response to pathogens in vivo, by generating mouse lines lacking the DC-SIGN homologues SIGNR1, SIGNR3, and SIGNR5. Resistance to Mycobacterium tuberculosis was impaired only in SIGNR3-deficient animals. SIGNR3 was expressed in lung phagocytes during infection, and interacted with M. tuberculosis bacilli and mycobacterial surface glycoconjugates to induce secretion of critical host defense inflammatory cytokines, including tumor necrosis factor (TNF). SIGNR3 signaling was dependent on an intracellular tyrosine-based motif and the tyrosine kinase Syk. Thus, the mouse DC-SIGN homologue SIGNR3 makes a unique contribution to protection of the host against a pulmonary bacterial pathogen.


Assuntos
Moléculas de Adesão Celular/fisiologia , Lectinas Tipo C/fisiologia , Receptores de Superfície Celular/fisiologia , Tuberculose/imunologia , Animais , Antígenos CD/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Glicoconjugados/metabolismo , Interleucina-6/biossíntese , Lipopolissacarídeos/metabolismo , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Transdução de Sinais , Receptor 2 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/biossíntese
12.
PLoS Pathog ; 4(11): e1000204, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19002241

RESUMO

Tuberculosis (TB) is characterized by a tight interplay between Mycobacterium tuberculosis and host cells within granulomas. These cellular aggregates restrict bacterial spreading, but do not kill all the bacilli, which can persist for years. In-depth investigation of M. tuberculosis interactions with granuloma-specific cell populations are needed to gain insight into mycobacterial persistence, and to better understand the physiopathology of the disease. We have analyzed the formation of foamy macrophages (FMs), a granuloma-specific cell population characterized by its high lipid content, and studied their interaction with the tubercle bacillus. Within our in vitro human granuloma model, M. tuberculosis long chain fatty acids, namely oxygenated mycolic acids (MA), triggered the differentiation of human monocyte-derived macrophages into FMs. In these cells, mycobacteria no longer replicated and switched to a dormant non-replicative state. Electron microscopy observation of M. tuberculosis-infected FMs showed that the mycobacteria-containing phagosomes migrate towards host cell lipid bodies (LB), a process which culminates with the engulfment of the bacillus into the lipid droplets and with the accumulation of lipids within the microbe. Altogether, our results suggest that oxygenated mycolic acids from M. tuberculosis play a crucial role in the differentiation of macrophages into FMs. These cells might constitute a reservoir used by the tubercle bacillus for long-term persistence within its human host, and could provide a relevant model for the screening of new antimicrobials against non-replicating persistent mycobacteria.


Assuntos
Diferenciação Celular , Células Espumosas/microbiologia , Granuloma/microbiologia , Macrófagos/microbiologia , Ácidos Micólicos , Tuberculose/microbiologia , Humanos , Lipídeos , Macrófagos/patologia , Macrófagos/ultraestrutura , Infecções por Mycobacterium/imunologia , Mycobacterium tuberculosis/fisiologia , Fagocitose , Tuberculose/imunologia
13.
Mol Microbiol ; 70(3): 762-74, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18808383

RESUMO

Mycobacterium tuberculosis and other pathogenic mycobacterial species produce large amounts of a glycogen-like alpha-glucan that represents the major polysaccharide of their outermost capsular layer. To determine the role of the surface-exposed glucan in the physiology and virulence of these bacteria, orthologues of the glg genes involved in the biosynthesis of glycogen in Escherichia coli were identified in M. tuberculosis H37Rv and inactivated by allelic replacement. Biochemical analyses of the mutants and complemented strains indicated that the synthesis of glucan and glycogen involves the alpha-1,4-glucosyltransferases Rv3032 and GlgA (Rv1212c), the ADP-glucose pyrophosphorylase GlgC (Rv1213) and the branching enzyme GlgB (Rv1326c). Disruption of glgC reduced by half the glucan and glycogen contents of M. tuberculosis, whereas the inactivation of glgA and Rv3032 affected the production of capsular glucan and glycogen, respectively. Attempts to disrupt Rv3032 in the glgA mutant were unsuccessful, suggesting that a functional copy of at least one of the two alpha-1,4-glucosyltransferases is required for growth. Importantly, the glgA mutant was impaired in its ability to persist in mice, suggesting a role for the capsular glucan in the persistence phase of infection. Unexpectedly, GlgB was found to be an essential enzyme.


Assuntos
Proteínas de Bactérias/metabolismo , Glucanos/biossíntese , Glicogênio/biossíntese , Mycobacterium tuberculosis/metabolismo , Tuberculose/microbiologia , Enzima Ramificadora de 1,4-alfa-Glucana/genética , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Animais , Proteínas de Bactérias/genética , Células Cultivadas , DNA Bacteriano/genética , Feminino , Técnicas de Inativação de Genes , Genes Bacterianos , Teste de Complementação Genética , Glucose-1-Fosfato Adenililtransferase/genética , Glucose-1-Fosfato Adenililtransferase/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Mycobacterium tuberculosis/genética
14.
Cell Microbiol ; 9(5): 1252-61, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17223928

RESUMO

Adherent-invasive Escherichia coli (AIEC) have been shown to be highly associated with ileal Crohn's disease (CD). AIEC survive within infected macrophages, residing within the phagolysosomal compartment where they take advantage of the low pH to replicate extensively. We investigated whether, like the tuberculous bacillus which also persists within macrophages, AIEC LF82 induces the formation of granulomas, which are a common histopathological feature of CD. For this purpose, we have taken advantage of an in vitro model of human granulomas that we recently developed, based on blood-derived mononuclear cells. We demonstrated that AIEC LF82 induces aggregation of infected macrophages, fusion of some of them to form multinucleated giant cells and subsequent recruitment of lymphocytes. Light microscopy and scanning electron microscopy analysis of the cell aggregates confirmed their granuloma features. This was further confirmed by histological analysis of granuloma sections. Noteworthy, this phenomenon can be reproduced by soluble protein extracts of AIEC LF82 coated onto beads. Although the cell aggregates not completely mimic natural CD-associated granulomas, they are very similar to early stages of epithelioid granulomas.


Assuntos
Doença de Crohn/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/crescimento & desenvolvimento , Granuloma/microbiologia , Aderência Bacteriana , Agregação Celular , Células Cultivadas , Escherichia coli/ultraestrutura , Humanos , Macrófagos/citologia , Macrófagos/microbiologia , Microscopia Eletrônica de Varredura , Monócitos/citologia , Monócitos/microbiologia , Monócitos/ultraestrutura , Fagossomos/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...