Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biomed Sci ; 31(1): 18, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38287360

RESUMO

BACKGROUND: Mycobacterium abscessus, a fast-growing non-tuberculous mycobacterium, is an emerging opportunistic pathogen responsible for chronic bronchopulmonary infections in people with respiratory diseases such as cystic fibrosis (CF). Due to its intrinsic polyresistance to a wide range of antibiotics, most treatments for M. abscessus pulmonary infections are poorly effective. In this context, antimicrobial peptides (AMPs) active against bacterial strains and less prompt to cause resistance, represent a good alternative to conventional antibiotics. Herein, we evaluated the effect of three arenicin isoforms, possessing two or four Cysteines involved in one (Ar-1, Ar-2) or two disulfide bonds (Ar-3), on the in vitro growth of M. abscessus. METHODS: The respective disulfide-free AMPs, were built by replacing the Cysteines with alpha-amino-n-butyric acid (Abu) residue. We evaluated the efficiency of the eight arenicin derivatives through their antimicrobial activity against M. abscessus strains, their cytotoxicity towards human cell lines, and their hemolytic activity on human erythrocytes. The mechanism of action of the Ar-1 peptide was further investigated through membrane permeabilization assay, electron microscopy, lipid insertion assay via surface pressure measurement, and the induction of resistance assay. RESULTS: Our results demonstrated that Ar-1 was the safest peptide with no toxicity towards human cells and no hemolytic activity, and the most active against M. abscessus growth. Ar-1 acts by insertion into mycobacterial lipids, resulting in a rapid membranolytic effect that kills M. abscessus without induction of resistance. CONCLUSION: Overall, the present study emphasized Ar-1 as a potential new alternative to conventional antibiotics in the treatment of CF-associated bacterial infection related to M. abscessus.


Assuntos
Fibrose Cística , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Poliestirenos , Humanos , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Antibacterianos/farmacologia , Fibrose Cística/tratamento farmacológico , Fibrose Cística/microbiologia , Peptídeos/farmacologia , Testes de Sensibilidade Microbiana
2.
FEBS J ; 290(6): 1563-1582, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36197115

RESUMO

A hallmark of Mycobacterium tuberculosis (M. tb), the aetiologic agent of tuberculosis, is its ability to metabolise host-derived lipids. However, the enzymes and mechanisms underlying such metabolism are still largely unknown. We previously reported that the Cyclophostin & Cyclipostins (CyC) analogues, a new family of potent antimycobacterial molecules, react specifically and covalently with (Ser/Cys)-based enzymes mostly involved in bacterial lipid metabolism. Here, we report the synthesis of new CyC alkyne-containing inhibitors (CyCyne ) and their use for the direct fishing of target proteins in M. tb culture via bio-orthogonal click-chemistry activity-based protein profiling (CC-ABPP). This approach led to the capture and identification of a variety of enzymes, and many of them involved in lipid or steroid metabolisms. One of the captured enzymes, HsaD (Rv3569c), is required for the survival of M. tb within macrophages and is thus a potential therapeutic target. This prompted us to further explore and validate, through a combination of biochemical and structural approaches, the specificity of HsaD inhibition by the CyC analogues. We confirmed that the CyC bind covalently to the catalytic Ser114 residue, leading to a total loss of enzyme activity. These data were supported by the X-ray structures of four HsaD-CyC complexes, obtained at resolutions between 1.6 and 2.6 Å. The identification of mycobacterial enzymes directly captured by the CyCyne probes through CC-ABPP paves the way to better understand and potentially target key players at crucial stages of the bacilli life cycle.


Assuntos
Antituberculosos , Proteínas de Bactérias , Hidrolases , Simulação de Acoplamento Molecular , Mycobacterium tuberculosis , Compostos Organofosforados , Humanos , Antituberculosos/síntese química , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Macrófagos/microbiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Tuberculose/tratamento farmacológico , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Compostos Organofosforados/química , Cristalografia por Raios X , Hidrolases/antagonistas & inibidores , Hidrolases/química , Simulação por Computador
3.
ACS Infect Dis ; 8(12): 2564-2578, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36379042

RESUMO

Patients with cystic fibrosis (CF) have a significantly higher risk of acquiring nontuberculous mycobacteria infections, predominantly due to Mycobacterium abscessus, than the healthy population. Because M. abscessus infections are a major cause of clinical decline and morbidity in CF patients, improving treatment and the detection of this mycobacterium in the context of a polymicrobial culture represents a critical component to better manage patient care. We report here the synthesis of fluorescent Dansyl derivatives of four active cyclipostins and cyclophostin analogues (CyCs) and provide new insights regarding the CyC's lack of activity against Gram-negative and Gram-positive bacteria, and above all into their mode of action against intramacrophagic M. abscessus cells. Our results pointed out that the intracellularly active CyC accumulate in acidic compartments within macrophage cells, that this accumulation appears to be essential for their delivery to mycobacteria-containing phagosomes, and consequently, for their antimicrobial effect against intracellular replicating M. abscessus, and that modification of such intracellular localization via disruption of endolysosomal pH strongly affects the CyC accumulation and efficacy. Moreover, we discovered that these fluorescent compounds could become efficient probes to specifically label mycobacterial species with high sensitivity, including M. abscessus in the presence several other pathogens like Pseudomonas aeruginosa and Staphylococcus aureus. Collectively, all present and previous data emphasized the therapeutic potential of unlabeled CyCs and the attractiveness of the fluorescent CyC as a potential new efficient diagnostic tool to be exploited in future diagnostic developments against mycobacterial-related infections, especially against M. abscessus.

4.
Bioorg Med Chem Lett ; 64: 128692, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35307568

RESUMO

With the aim to discover new antituberculous molecules, three novel series of 23 hydroxamic acids, 13 hydrazides, and 9O-alkyl/O-acyl protected hydroxamic acid derivatives have been synthesized, and fully characterized by spectral 1H NMR, 13C NMR, HRMS) analysis. These compounds were further biologically screened for their in vitro antibacterial activities against three pathogenic mycobacteria - M. abscessus S and R, M. marinum, and M. tuberculosis - as well as for their toxicity towards murine macrophages by the resazurin microtiter assay (REMA). Among the 45 derivatives, 17 compounds (3 hydroxamic acids, 9 hydrazides, and 5O-alkyl/O-acyl protected hydroxamic acids) were nontoxic against murine macrophages. When tested for their antibacterial activity, hydroxamic acid 9 h was found to be the most potent inhibitor against M. abscessus S and R only. Regarding hydrazide series, only 7h was active against M. abscessus R, M. marinum and M. tuberculosis; while the O-acyl protected hydroxamic acid derivatives 14d and 15d displayed promising antibacterial activity against both M. marinum and M. tuberculosis. Since such hydroxamic- and hydrazide-chelating groups have been reported to impair the activity of the peptide deformylase, in silico molecular docking studies in M. tuberculosis peptide deformylase enzyme active site were further performed with 7h in order to predict the possible interaction mode and binding energy of this molecule at the molecular level.


Assuntos
Ácidos Hidroxâmicos , Mycobacterium tuberculosis , Animais , Antibacterianos/química , Hidrazinas/farmacologia , Ácidos Hidroxâmicos/química , Camundongos , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade
5.
Virulence ; 12(1): 1438-1451, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34107844

RESUMO

Mycobacterium ulcerans is the causal agent of Buruli ulcer, a chronic infectious disease and the third most common mycobacterial disease worldwide. Without early treatment, M. ulcerans provokes massive skin ulcers, caused by the mycolactone toxin, its main virulence factor. However, spontaneous healing may occur in Buruli ulcer patients several months or years after the disease onset. We have shown, in an original mouse model, that bacterial load remains high and viable in spontaneously healed tissues, with a switch of M. ulcerans to low levels of mycolactone production, adapting its strategy to survive in such a hostile environment. This original model offers the possibility to investigate the regulation of mycolactone production, by using an RNA-seq strategy to study bacterial adaptation during mouse infection. Pathway analysis and characterization of the tissue environment showed that the bacillus adapted to its new environment by modifying its metabolic activity and switching nutrient sources. Thus, M. ulcerans ensures its survival in healing tissues by reducing its secondary metabolism, leading to an inhibition of mycolactone synthesis. These findings shed new light on mycolactone regulation and pave the way for new therapeutic strategies.


Assuntos
Úlcera de Buruli , Macrolídeos/metabolismo , Infecções por Mycobacterium , Mycobacterium ulcerans , Adaptação Biológica , Animais , Úlcera de Buruli/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Infecções por Mycobacterium/microbiologia , Mycobacterium ulcerans/genética
6.
FEMS Microbiol Rev ; 45(6)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34036305

RESUMO

Mycobacterial species, including Mycobacterium tuberculosis, rely on lipids to survive and chronically persist within their hosts. Upon infection, opportunistic and strict pathogenic mycobacteria exploit metabolic pathways to import and process host-derived free fatty acids, subsequently stored as triacylglycerols in the form of intrabacterial lipid inclusions (ILI). Under nutrient-limiting conditions, ILI constitute a critical source of energy that fuels the carbon requirements and maintain redox homeostasis, promoting bacterial survival for extensive periods of time. In addition to their basic metabolic functions, these organelles display multiple other biological properties, emphasizing their central role in the mycobacterial life cycle. However, despite their importance, the dynamics of ILI metabolism and their contribution to mycobacterial adaptation/survival in the context of infection has not been thoroughly documented. Herein, we provide an overview of the historical ILI discoveries, their characterization and current knowledge regarding the microenvironmental stimuli conveying ILI formation, storage and degradation. We also review new biological systems to monitor the dynamics of ILI metabolism in extra- and intracellular mycobacteria and describe major molecular actors in triacylglycerol biosynthesis, maintenance and breakdown. Finally, emerging concepts regarding the role of ILI in mycobacterial survival, persistence, reactivation, antibiotic susceptibility and inter-individual transmission are also discussed.


Assuntos
Mycobacterium tuberculosis , Lipídeos , Triglicerídeos
7.
Sci Rep ; 9(1): 8667, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31209261

RESUMO

Mycobacteria share with other actinomycetes the ability to produce large quantities of triacylglycerol (TAG), which accumulate as intracytoplasmic lipid inclusions (ILI) also known as lipid droplets (LD). Mycobacterium tuberculosis (M. tb), the etiologic agent of tuberculosis, acquires fatty acids from the human host which are utilized to synthesize TAG, subsequently stored in the form of ILI to meet the carbon and nutrient requirements of the bacterium during long periods of persistence. However, environmental factors governing mycobacterial ILI formation and degradation remain poorly understood. Herein, we demonstrated that in the absence of host cells, carbon excess and nitrogen starvation promote TAG accumulation in the form of ILI in M. smegmatis and M. abscessus, used as surrogate species of M. tb. Based on these findings, we developed a simple and reversible in vitro model to regulate ILI biosynthesis and hydrolysis in mycobacteria. We also showed that TAG formation is tgs1 dependent and that lipolytic enzymes mediate TAG breakdown. Moreover, we confirmed that the nitrogen-deprived and ILI-rich phenotype was associated with an increased tolerance towards several drugs used for treating mycobacterial infections. Importantly, we showed that the presence of ILI substantially enhanced the bacterial burden and granuloma abundance in zebrafish embryos infected with lipid-rich M. abscessus as compared to embryos infected with lipid-poor M. abscessus, suggesting that ILI are actively contributing to mycobacterial virulence and pathogenesis.


Assuntos
Antituberculosos/farmacologia , Infecções por Mycobacterium não Tuberculosas/metabolismo , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium smegmatis/efeitos dos fármacos , Nitrogênio/deficiência , Triglicerídeos/biossíntese , Animais , Animais Geneticamente Modificados , Carbono/metabolismo , Tolerância a Medicamentos , Embrião não Mamífero , Ácidos Graxos/metabolismo , Humanos , Isoniazida/farmacologia , Ligases/genética , Ligases/metabolismo , Gotículas Lipídicas/metabolismo , Lipólise , Longevidade/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/mortalidade , Mycobacterium abscessus/metabolismo , Mycobacterium abscessus/patogenicidade , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Rifampina/farmacologia , Virulência , Peixe-Zebra
8.
FEBS J ; 286(16): 3164-3181, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31034693

RESUMO

The Mycobacterium tuberculosis LipY protein, a prototype of the proline-glutamic acid (PE) family, exhibits a triacylglycerol (TAG) hydrolase activity that contributes to host cell lipid degradation and persistence of the bacilli. LipY is found either as a full-length intracytosolic form or as a mature extracellular form lacking the N-terminal PE domain. Even though the contribution of the extracellular form in TAG consumption has been partly elucidated, very little information is available regarding the potential interactions of either full-length LipY with the cytoplasmic membrane, or mature form LipY with the outer membrane. Herein, several LipY variants truncated in their N-terminal domain were produced and biochemically characterized in lipid-protein interaction assays, using the monomolecular film technique and FTIR. Comparison of the catalytic activities of these recombinant proteins showed that LipY∆149, corresponding to the extracellular form of LipY lacking the PE domain, is more active than the full-length protein. This confirms previous studies reporting that the PE domain negatively modulates the TAG hydrolase activity of LipY. Lipid-protein interaction studies indicate that the PE domain anchors LipY onto membrane lipids. Consistent with these findings, we show that LipY∆149 is loosely associated with the mycobacterial cell wall, and that this interaction is mediated by the sole lipase domain. Overall, our results bring new information regarding the molecular mechanisms by which LipY either binds and hydrolyses host cell lipids or degrades TAG, the major source of lipids within mycobacterial intracytosolic lipid inclusions.


Assuntos
Proteínas de Bactérias/genética , Hidrolases de Éster Carboxílico/genética , Metabolismo dos Lipídeos/genética , Lipídeos de Membrana/genética , Mycobacterium tuberculosis/genética , Fatores de Virulência/genética , Proteínas de Bactérias/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Catálise , Parede Celular/genética , Parede Celular/metabolismo , Lipase/genética , Lipídeos de Membrana/metabolismo , Mycobacterium tuberculosis/metabolismo , Ligação Proteica/genética , Domínios Proteicos/genética , Triglicerídeos/genética , Triglicerídeos/metabolismo , Fatores de Virulência/metabolismo
9.
Biosci Rep ; 38(6)2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30487163

RESUMO

Tuberculosis caused by Mycobacterium tuberculosis is currently one of the leading causes of death from an infectious agent. The main difficulties encountered in eradicating this bacteria are mainly related to (i) a very complex lipid composition of the bacillus cell wall, (ii) its ability to hide from the immune system inside the granulomas, and (iii) the increasing number of resistant strains. In this context, we were interested in the Rv0646c (lipGMTB ) gene located upstream to the mmaA cluster which is described as being crucial for the production of cell wall components and required for the bacilli adaptation and survival in mouse macrophages. Using biochemical experiments combined with the construction of deletion and overexpression mutant strains in Mycobacterium smegmatis, we found that LipGMTB is a cytoplasmic membrane-associated enzyme that displays both phospholipase and thioesterase activities. Overproduction of LipGMTB decreases the glycopeptidolipids (GPL) level concomitantly to an increase in phosphatidylinositol (PI) which is the precursor of the PI mannoside (PIM), an essential lipid component of the bacterial cell wall. Conversely, deletion of the lipGMS gene in M. smegmatis leads to an overproduction of GPL, and subsequently decreases the strain susceptibility to various antibiotics. All these findings demonstrate that LipG is involved in cell envelope biosynthesis/remodeling, and consequently this enzyme may thus play an important role in mycobacterial physiology.


Assuntos
Parede Celular/enzimologia , Glicopeptídeos/genética , Fosfolipases/genética , Tuberculose/microbiologia , Animais , Antibacterianos/farmacologia , Parede Celular/química , Glicolipídeos/química , Glicolipídeos/genética , Glicopeptídeos/química , Humanos , Macrófagos/enzimologia , Camundongos , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/enzimologia , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/patogenicidade , Fosfatidilinositóis/química , Fosfatidilinositóis/metabolismo , Fosfolipases/química , Tuberculose/enzimologia
10.
Infect Immun ; 86(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29986895

RESUMO

Within tuberculous granulomas, a subpopulation of Mycobacterium tuberculosis resides inside foamy macrophages (FM) that contain abundant cytoplasmic lipid bodies (LB) filled with triacylglycerol (TAG). Upon fusion of LB with M. tuberculosis-containing phagosomes, TAG is hydrolyzed and reprocessed by the bacteria into their own lipids, which accumulate as intracytosolic lipid inclusions (ILI). This phenomenon is driven by many mycobacterial lipases, among which LipY participates in the hydrolysis of host and bacterial TAG. However, the functional contribution of LipY's PE domain to TAG hydrolysis remains unclear. Here, enzymatic studies were performed to compare the lipolytic activities of recombinant LipY and its truncated variant lacking the N-terminal PE domain, LipY(ΔPE). Complementarily, an FM model was used where bone marrow-derived mouse macrophages were infected with M. bovis BCG strains either overexpressing LipY or LipY(ΔPE) or carrying a lipY deletion mutation prior to being exposed to TAG-rich very-low-density lipoprotein (VLDL). Results indicate that truncation of the PE domain correlates with increased TAG hydrolase activity. Quantitative electron microscopy analyses showed that (i) in the presence of lipase inhibitors, large ILI (ILI+3) were not formed because of an absence of LB due to inhibition of VLDL-TAG hydrolysis or inhibition of LB-neutral lipid hydrolysis by mycobacterial lipases, (ii) ILI+3 profiles in the strain overexpressing LipY(ΔPE) were reduced, and (iii) the number of ILI+3 profiles in the ΔlipY mutant was reduced by 50%. Overall, these results delineate the role of LipY and its PE domain in host and mycobacterial lipid consumption and show that additional mycobacterial lipases take part in these processes.


Assuntos
Proteínas de Bactérias/química , Hidrolases de Éster Carboxílico/química , Metabolismo dos Lipídeos , Macrófagos/microbiologia , Macrófagos/fisiologia , Triglicerídeos/metabolismo , Fatores de Virulência/química , Animais , Proteínas de Bactérias/genética , Hidrolases de Éster Carboxílico/genética , Domínio Catalítico , Células Cultivadas , Feminino , Lipase/metabolismo , Lipoproteínas VLDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Mycobacterium bovis , Estrutura Terciária de Proteína , Tuberculose/microbiologia , Fatores de Virulência/genética
11.
Artigo em Inglês | MEDLINE | ID: mdl-28567368

RESUMO

Adipose tissues were shown to host Mycobacterium tuberculosis which is persisting inside mature adipocytes. It remains unknown whether this holds true for Mycobacterium canettii, a rare representative of the M. tuberculosis complex responsible for lymphatic and pulmonary tuberculosis. Here, we infected primary murine white and brown pre-adipocytes and murine 3T3-L1 pre-adipocytes and mature adipocytes with M. canettii and M. tuberculosis as a positive control. Both mycobacteria were able to infect 18-22% of challenged primary murine pre-adipocytes; and to replicate within these cells during a 7-day experiment with the intracellular inoculums being significantly higher in brown than in white pre-adipocytes for M. canettii (p = 0.02) and M. tuberculosis (p = 0.03). Further in-vitro infection of 3T3-L1 mature adipocytes yielded 9% of infected cells by M. canettii and 17% of infected cells by M. tuberculosis (p = 0.001). Interestingly, M. canettii replicated and accumulated intra-cytosolic lipid inclusions within mature adipocytes over a 12-day experiment; while M. tuberculosis stopped replicating at day 3 post-infection. These results indicate that brown pre-adipocytes could be one of the potential targets for M. tuberculosis complex mycobacteria; and illustrate differential outcome of M. tuberculosis complex mycobacteria into adipose tissues. While white adipose tissue is an unlikely sanctuary for M. canettii, it is still an open question whether M. canettii and M. tuberculosis could persist in brown adipose tissues.


Assuntos
Tecido Adiposo/microbiologia , Tecido Adiposo/patologia , Mycobacterium/patogenicidade , Tuberculose/microbiologia , Células 3T3-L1 , Adipócitos/microbiologia , Adipócitos/patologia , Tecido Adiposo/diagnóstico por imagem , Animais , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/classificação , Mycobacterium tuberculosis/patogenicidade , Tuberculose/diagnóstico por imagem
12.
Sci Rep ; 7: 43682, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272507

RESUMO

Post-translational modifications (PTMs) are essential processes conditioning the biophysical properties and biological activities of the vast majority of mature proteins. However, occurrence of several distinct PTMs on a same protein dramatically increases its molecular diversity. The comprehensive understanding of the functionalities resulting from any particular PTM association requires a highly challenging full structural description of the PTM combinations. Here, we report the in-depth exploration of the natural structural diversity of the M. tuberculosis (Mtb) virulence associated 19 kDa lipoglycoprotein antigen (LpqH) using intact protein high-resolution mass spectrometry (HR-MS) coupled to liquid chromatography. Combined top-down and bottom-up HR-MS analyses of the purified Mtb LpqH protein allow, for the first time, to uncover a complex repertoire of about 130 molecular species resulting from the intrinsically heterogeneous combination of lipidation and glycosylation together with some truncations. Direct view on the co-occurring PTMs stoichiometry reveals the presence of functionally distinct LpqH lipidation states and indicates that glycosylation is independent from lipidation. This work allowed the identification of a novel unsuspected phosphorylated form of the unprocessed preprolipoglycoprotein totally absent from the current lipoglycoprotein biogenesis pathway and providing new insights into the biogenesis and functional determinants of the mycobacterial lipoglycoprotein interacting with the host immune PRRs.


Assuntos
Proteínas de Bactérias/metabolismo , Glicoproteínas/biossíntese , Lipoproteínas/biossíntese , Mycobacterium tuberculosis/metabolismo , Proteoma , Proteômica , Acetilação , Sequência de Aminoácidos , Proteínas de Bactérias/imunologia , Cromatografia Líquida , Modelos Biológicos , Mycobacterium tuberculosis/imunologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteômica/métodos , Espectrometria de Massas em Tandem
13.
Artigo em Inglês | MEDLINE | ID: mdl-27774438

RESUMO

Despite a slight decline since 2014, tuberculosis (TB) remains the major deadly infectious disease worldwide with about 1.5 million deaths each year and with about one-third of the population being latently infected with Mycobacterium tuberculosis, the etiologic agent of TB. During primo-infection, the recruitment of immune cells leads to the formation of highly organized granulomas. Among the different cells, one outstanding subpopulation is the foamy macrophage (FM), characterized by the abundance of triacylglycerol-rich lipid bodies (LB). M. tuberculosis can reside in FM, where it acquires, from host LB, the neutral lipids which are subsequently processed and stored by the bacilli in the form of intracytosolic lipid inclusions (ILI). Although host LB can be viewed as a reservoir of nutrients for the pathogen during latency, the molecular mechanisms whereby intraphagosomal mycobacteria interact with LB and assimilate the LB-derived lipids are only beginning to be understood. Past studies have emphasized that these physiological processes are critical to the M. tuberculosis infectious-life cycle, for propagation of the infection, establishment of the dormancy state and reactivation of the disease. In recent years, several animal and cellular models have been developed with the aim of dissecting these complex processes and of determining the nature and contribution of their key players. Herein, we review some of the in vitro and in vivo models which allowed to gain significant insight into lipid accumulation and consumption in M. tuberculosis, two important events that are directly linked to pathogenicity, granuloma formation/maintenance and survival of the tubercle bacillus under non-replicative conditions. We also discuss the advantages and limitations of each model, hoping that this will serve as a guide for future investigations dedicated to persistence and innovative therapeutic approaches against TB.


Assuntos
Interações Hospedeiro-Patógeno , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Animais , Humanos , Modelos Teóricos
14.
Vaccine ; 33(18): 2118-24, 2015 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-25804706

RESUMO

BACKGROUND: Vaccine strategies represent one of the fighting answers against multiresistant bacteria in a number of clinical settings like cystic fibrosis (CF). Mycobacterium abscessus, an emerging CF pathogen, raises difficult therapeutic problems due to its intrinsic antibiotic multiresistance. METHODS: By reverse vaccinology, we identified M. abscessus phospholipase C (MA-PLC) as a potential vaccine target. We deciphered here the protective response generated by vaccination with plasmid DNA encoding the MA-PLC formulated with a tetra functional block copolymer 704, in CF (ΔF508) mice. Protection was tested against aerosolized smooth and rough (hypervirulent) variants of M. abscessus. RESULTS: MA-PLC DNA vaccination (days 0, 21, 42) elicited a strong antibody response. A significant protective effect was obtained against aerosolized M. abscessus (S variant) in ΔF508 mice, but not in wild-type FVB littermates; similar results were observed when: (i) challenging mice with the "hypervirulent" R variant, and; (ii) immunizing mice with purified MA-PLC protein. High IgG titers against MA-PLC protein were measured in CF patients with M. abscessus infection; interestingly, significant titers were also detected in CF patients positive for Pseudomonas aeruginosa versus P. aeruginosa-negative controls. CONCLUSIONS: MA-PLC DNA- and PLC protein-vaccinated mice cleared more rapidly M. abscessus than ß-galactosidase DNA- or PBS- vaccinated mice in the context of CF. PLCs could constitute interesting vaccine targets against common PLC-producing CF pathogens like P. aeruginosa.


Assuntos
Vacinas Bacterianas/imunologia , Fibrose Cística/complicações , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Micobactérias não Tuberculosas/imunologia , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Feminino , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Masculino , Camundongos , Infecções por Mycobacterium não Tuberculosas/complicações , Micobactérias não Tuberculosas/enzimologia , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Fosfolipases Tipo C/administração & dosagem , Vacinas de DNA/administração & dosagem , beta-Galactosidase/genética , beta-Galactosidase/imunologia
15.
Infect Immun ; 83(2): 780-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25486995

RESUMO

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium involved in pulmonary and cutaneo-mucous infections worldwide, to which cystic fibrosis patients are exquisitely susceptible. The analysis of the genome sequence of M. abscessus showed that this bacterium is endowed with the metabolic pathways typically found in environmental microorganisms that come into contact with soil, plants, and aquatic environments, where free-living amoebae are frequently present. M. abscessus also contains several genes that are characteristically found only in pathogenic bacteria. One of them is MAB_0555, encoding a putative phospholipase C (PLC) that is absent from most other rapidly growing mycobacteria, including Mycobacterium chelonae and Mycobacterium smegmatis. Here, we report that purified recombinant M. abscessus PLC is highly cytotoxic to mouse macrophages, presumably due to hydrolysis of membrane phospholipids. We further showed by constructing and using an M. abscessus PLC knockout mutant that loss of PLC activity is deleterious to M. abscessus intracellular survival in amoebae. The importance of PLC is further supported by the fact that M. abscessus PLC was found to be expressed only in amoebae. Aerosol challenge of mice with M. abscessus strains that were precultured in amoebae enhanced M. abscessus lung infectivity relative to M. abscessus grown in broth culture. Our study underlines the importance of PLC for the virulence of M. abscessus. Despite the difficulties of isolating M. abscessus from environmental sources, our findings suggest that M. abscessus has evolved in close contact with environmental protozoa, which supports the argument that amoebae may contribute to the virulence of opportunistic mycobacteria.


Assuntos
Amoeba/fisiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium/patogenicidade , Fosfolipases Tipo C/fisiologia , Amoeba/microbiologia , Animais , Sequência de Bases , Células Cultivadas , Técnicas de Cocultura , Fibrose Cística/microbiologia , Técnicas de Inativação de Genes , Genoma Bacteriano/genética , Macrófagos/imunologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/enzimologia , Mycobacterium/genética , Infecções por Mycobacterium não Tuberculosas/microbiologia , Proteínas Recombinantes , Análise de Sequência de DNA , Fosfolipases Tipo C/genética , Fatores de Virulência/genética
16.
Food Funct ; 5(7): 1409-21, 2014 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24777447

RESUMO

CITREM is an emulsifier used in the food industry and contains citric acid esters of mono- and diglycerides (GCFE). It is generally recognized as safe but no publication on its digestibility under gastrointestinal conditions and impact on fat digestion was available. It was shown here that fatty acids are released from CITREM by gastric lipase, pancreatic lipase, pancreatic-lipase-related protein 2 and carboxyl ester hydrolase. A two-step in vitro digestion model mimicking lipolysis in the stomach and upper small intestine of term and preterm infants was then used to evaluate the digestibility of CITREM alone, CITREM-containing infant formula and fat emulsions, and isolated GCFE fractions. Overall, it was shown that fat digestion is not significantly changed by the presence of CITREM, and fatty acids contained in CITREM compounds are released to a large extent by lipases. Nevertheless, undigestible water-soluble compounds containing glycerol and citric acid units were identified, indicating that the ester bond between citric acid and glycerol is not fully hydrolyzed throughout the proposed digestion.


Assuntos
Citratos/metabolismo , Diglicerídeos/metabolismo , Emulsificantes/metabolismo , Ésteres/metabolismo , Fórmulas Infantis/química , Monoglicerídeos/metabolismo , Carboxilesterase/metabolismo , Digestão , Emulsões/química , Ácidos Graxos/metabolismo , Trato Gastrointestinal/enzimologia , Humanos , Lactente , Lipase/metabolismo , Lipólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA