Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
PLoS One ; 19(5): e0303516, 2024.
Article in English | MEDLINE | ID: mdl-38728330

ABSTRACT

Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.


Subject(s)
Succinates , Succinates/pharmacology , Succinates/metabolism , Humans , Hydrogen-Ion Concentration , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/growth & development , THP-1 Cells , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/metabolism
2.
Microbiol Spectr ; 10(1): e0132121, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019671

ABSTRACT

The aminobenzimidazole SPR719 targets DNA gyrase in Mycobacterium tuberculosis. The molecule acts as inhibitor of the enzyme's ATPase located on the Gyrase B subunit of the tetrameric Gyrase A2B2 protein. SPR719 is also active against non-tuberculous mycobacteria (NTM) and recently entered clinical development for lung disease caused by these bacteria. Resistance against SPR719 in NTM has not been characterized. Here, we determined spontaneous in vitro resistance frequencies in single step resistance development studies, MICs of resistant strains, and resistance associated DNA sequence polymorphisms in two major NTM pathogens Mycobacterium avium and Mycobacterium abscessus. A low-frequency resistance (10-8/CFU) was associated with missense mutations in the ATPase domain of the Gyrase B subunit in both bacteria, consistent with inhibition of DNA gyrase as the mechanism of action of SPR719 against NTM. For M. abscessus, but not for M. avium, a second, high-frequency (10-6/CFU) resistance mechanism was observed. High-frequency SPR719 resistance was associated with frameshift mutations in the transcriptional repressor MAB_4384 previously shown to regulate expression of the drug efflux pump system MmpS5/MmpL5. Our results confirm DNA gyrase as target of SPR719 in NTM and reveal differential resistance development in the two NTM species, with M. abscessus displaying high-frequency indirect resistance possibly involving drug efflux. IMPORTANCE Clinical emergence of resistance to new antibiotics affects their utility. Characterization of in vitro resistance is a first step in the profiling of resistance properties of novel drug candidates. Here, we characterized in vitro resistance against SPR719, a drug candidate for the treatment of lung disease caused by non-tuberculous mycobacteria (NTM). The identified resistance associated mutations and the observed differential resistance behavior of the two characterized NTM species provide a basis for follow-up studies of resistance in vivo to further inform clinical development of SPR719.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/drug effects , Mycobacterium avium/drug effects , Topoisomerase II Inhibitors/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzimidazoles/pharmacology , DNA Gyrase/genetics , DNA Gyrase/metabolism , Drug Resistance, Bacterial , Humans , Microbial Sensitivity Tests , Mutation , Mycobacterium abscessus/enzymology , Mycobacterium abscessus/genetics , Mycobacterium abscessus/growth & development , Mycobacterium avium/enzymology , Mycobacterium avium/genetics , Mycobacterium avium/growth & development
3.
Nat Commun ; 12(1): 1606, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707445

ABSTRACT

Tuberculosis is a chronic disease that displays several features commonly associated with biofilm-associated infections: immune system evasion, antibiotic treatment failures, and recurrence of infection. However, although Mycobacterium tuberculosis (Mtb) can form cellulose-containing biofilms in vitro, it remains unclear whether biofilms are formed during infection in vivo. Here, we demonstrate the formation of Mtb biofilms in animal models of infection and in patients, and that biofilm formation can contribute to drug tolerance. First, we show that cellulose is also a structural component of the extracellular matrix of in vitro biofilms of fast and slow-growing nontuberculous mycobacteria. Then, we use cellulose as a biomarker to detect Mtb biofilms in the lungs of experimentally infected mice and non-human primates, as well as in lung tissue sections obtained from patients with tuberculosis. Mtb strains defective in biofilm formation are attenuated for survival in mice, suggesting that biofilms protect bacilli from the host immune system. Furthermore, the administration of nebulized cellulase enhances the antimycobacterial activity of isoniazid and rifampicin in infected mice, supporting a role for biofilms in phenotypic drug tolerance. Our findings thus indicate that Mtb biofilms are relevant to human tuberculosis.


Subject(s)
Biofilms/growth & development , Cellulose/metabolism , Drug Resistance, Multiple, Bacterial/physiology , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Animals , Cellulase/pharmacology , Disease Models, Animal , Drug Synergism , Humans , Isoniazid/pharmacology , Mice , Mice, Inbred C57BL , Mycobacterium abscessus/growth & development , Mycobacterium avium/growth & development , Mycobacterium fortuitum/growth & development , Mycobacterium tuberculosis/drug effects , Rifampin/pharmacology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/pathology
4.
Int J Mol Sci ; 23(1)2021 Dec 27.
Article in English | MEDLINE | ID: mdl-35008695

ABSTRACT

During infections, the host redistributes iron in order to starve pathogens from this nutrient. Several proteins are involved in iron absorption, transport, and storage. Ferritin is the most important iron storage protein. It is composed of variable proportions of two peptides, the L- and H-ferritins (FTL and FTH). We previously showed that macrophages increase their expression of FTH1 when they are infected in vitro with Mycobacterium avium, without a significant increase in FTL. In this work, we investigated the role of macrophage FTH1 in M. avium infection in vivo. We found that mice deficient in FTH1 in myeloid cells are more resistant to M. avium infection, presenting lower bacterial loads and lower levels of proinflammatory cytokines than wild-type littermates, due to the lower levels of available iron in the tissues. Importantly, we also found that FTH1 produced by myeloid cells in response to infection may be found in circulation and that it plays a key role in iron redistribution. Specifically, in the absence of FTH1 in myeloid cells, increased expression of ferroportin is observed in liver granulomas and increased iron accumulation occurs in hepatocytes. These results highlight the importance of FTH1 expression in myeloid cells for iron redistribution during infection.


Subject(s)
Blood Circulation , Ferritins/blood , Iron/metabolism , Liver/metabolism , Mycobacterium Infections/blood , Myeloid Cells/metabolism , Animals , Cation Transport Proteins/metabolism , Ferritins/deficiency , Gene Expression Regulation , Inflammation/pathology , Iron Deficiencies/blood , Iron Deficiencies/metabolism , Iron Overload/blood , Iron Overload/metabolism , Mice , Mycobacterium Infections/genetics , Mycobacterium avium/growth & development , Mycobacterium avium/physiology
5.
Microbes Infect ; 22(10): 567-575, 2020.
Article in English | MEDLINE | ID: mdl-32882411

ABSTRACT

Mycobacterium avium complex is a causative organism for refractory diseases. In this study, we examined the effects of N-acetyl-cysteine on M. avium infection in vitro and in vivo. N-acetyl-cysteine treatment suppressed the growth of M. avium in A549 cells in a concentration-dependent manner. This effect was related to the induction of the antibacterial peptide human ß-defensin-2. In a mouse model, N-acetyl-cysteine treatment significantly reduced the number of bacteria in the lungs and induced murine ß-defensin-3. In interleukin-17-deficient mice, the effects of N-acetyl-cysteine disappeared, indicating that these mechanisms may be mediated by interleukin-17. Moreover, an additional reduction in bacterial load was observed in mice administered N-acetyl-cysteine in combination with clarithromycin. Our findings demonstrate the potent antimycobacterial effects of N-acetyl-cysteine against M. avium by inducing antimicrobial peptide, suggesting that N-acetyl-cysteine may have applications as an alternative to classical treatment regimens.


Subject(s)
Acetylcysteine/pharmacology , Antitubercular Agents/pharmacology , Mycobacterium avium-intracellulare Infection/drug therapy , Mycobacterium avium/drug effects , beta-Defensins/metabolism , A549 Cells , Acetylcysteine/therapeutic use , Animals , Antitubercular Agents/therapeutic use , Bacterial Load/drug effects , Disease Models, Animal , Humans , Interleukin-17/metabolism , Lung/drug effects , Lung/microbiology , Mice , Mycobacterium avium/growth & development , Mycobacterium avium-intracellulare Infection/metabolism , Mycobacterium avium-intracellulare Infection/microbiology , RAW 264.7 Cells , Signal Transduction
6.
Molecules ; 25(10)2020 May 12.
Article in English | MEDLINE | ID: mdl-32408517

ABSTRACT

Based on the isosterism concept, we have designed and synthesized homologous N-alkyl-2-[4-(trifluoromethyl)benzoyl]hydrazine-1-carboxamides (from C1 to C18) as potential antimicrobial agents and enzyme inhibitors. They were obtained from 4-(trifluoromethyl)benzohydrazide by three synthetic approaches and characterized by spectral methods. The derivatives were screened for their inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) via Ellman's method. All the hydrazinecarboxamides revealed a moderate inhibition of both AChE and BuChE, with IC50 values of 27.04-106.75 µM and 58.01-277.48 µM, respectively. Some compounds exhibited lower IC50 for AChE than the clinically used drug rivastigmine. N-Tridecyl/pentadecyl-2-[4-(trifluoromethyl)benzoyl]hydrazine-1-carboxamides were identified as the most potent and selective inhibitors of AChE. For inhibition of BuChE, alkyl chain lengths from C5 to C7 are optimal substituents. Based on molecular docking study, the compounds may work as non-covalent inhibitors that are placed in a close proximity to the active site triad. The compounds were evaluated against Mycobacterium tuberculosis H37Rv and nontuberculous mycobacteria (M. avium, M. kansasii). Reflecting these results, we prepared additional analogues of the most active carboxamide (n-hexyl derivative 2f). N-Hexyl-5-[4-(trifluoromethyl)phenyl]-1,3,4-oxadiazol-2-amine (4) exhibited the lowest minimum inhibitory concentrations within this study (MIC ≥ 62.5 µM), however, this activity is mild. All the compounds avoided cytostatic properties on two eukaryotic cell lines (HepG2, MonoMac6).


Subject(s)
Acetylcholinesterase/metabolism , Anti-Infective Agents , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors , Imidazoles , Mycobacterium avium/growth & development , Mycobacterium kansasii/growth & development , Mycobacterium tuberculosis/growth & development , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , GPI-Linked Proteins/metabolism , Hep G2 Cells , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology
8.
Macromol Biosci ; 19(12): e1900254, 2019 12.
Article in English | MEDLINE | ID: mdl-31747130

ABSTRACT

Polycations, mimicking activity of antibacterial peptides, belong to an important class of molecules investigated as a support or as an alternative to antibiotics. In this work, studies of modified linear amphiphilic statistical polymethyloxazoline (PMOX) and polyethyleneimine copolymers (PMOX_PEI) series are presented. Variation of PEI content in the structure results in controllable changes of polymeric aggregates zeta potential. The structure with the highest positive charge shows the best antimicrobial activity, well visible in tests against model Gram-positive and Gram-negative bacteria, fungi, and mycobacterium strains. The polymer toxicity is evaluated with MTT and hemolysis assay as a reference. Quartz crystal microbalance (QCM-D) is used to investigate interaction between polycations and a model lipid membrane. Polymer activity correlates well with molecular structure, showing that amphiphilic component is altering polymer behavior in contact with the lipid bilayer.


Subject(s)
Anti-Infective Agents/pharmacology , Lipid Bilayers/chemistry , Polyamines/pharmacology , Polyethyleneimine/pharmacology , Anti-Infective Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Candida albicans/drug effects , Candida albicans/growth & development , Erythrocytes/drug effects , Escherichia coli/drug effects , Escherichia coli/growth & development , Hemolysis/drug effects , Humans , Microbial Sensitivity Tests , Molecular Mimicry , Molecular Structure , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium bovis/drug effects , Mycobacterium bovis/growth & development , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/growth & development , Polyamines/chemical synthesis , Polyelectrolytes/chemistry , Polyethyleneimine/chemical synthesis , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Static Electricity , Structure-Activity Relationship
9.
Acta Microbiol Immunol Hung ; 65(3): 405-418, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30024267

ABSTRACT

The aim of this study was to determine the effect of mycobacterial proteins on mycobacterial biofilm formation and growth processes. We separated growth-affecting proteins (GEPs) from wild type of Mycobacterium bovis and ATCC strain of Mycobacterium avium subsp. avium. Our results showed that these mycobacteria-secreted GEPs are involved in biofilm formation, growth stimulatory, and inhibitory processes. Our findings suggest that GEP stimulated M. avium subsp. avium growth in vitro. Stimulation process was observed in mycobacteria affected with GEP extracted from M. avium subsp. avium. We found that both GEPs inhibited the growth of the M. bovis. Optical density measurement and visual analysis confirm that GEP plays an important role in biofilm formation process. Most of M. bovis GEP are associated with the type VII secretion and general secretion pathways. Our results contribute to a better understanding of the mechanisms underlying mycobacterial biofilm formation and growth-affecting processes and better characterization of mycobacterial proteins and their functions. It is noteworthy that this finding represents the first demonstration of GEP-mediated growth effects on a solid and liquid medium.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Mycobacterium avium/physiology , Mycobacterium bovis/physiology , Bacterial Proteins/genetics , Humans , Mycobacterium avium/genetics , Mycobacterium avium/growth & development , Mycobacterium bovis/genetics , Mycobacterium bovis/growth & development
10.
J Infect Dis ; 218(2): 291-299, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29471363

ABSTRACT

Lysyl-phosphatidylglycerol is one of the components of the mycobacterial membrane that contributes to the resistance to cationic antimicrobial peptides, a host-induced frontline defense against invading pathogens. Its production is catalyzed by LysX, a bifunctional protein with lysyl transferase and lysyl transfer RNA synthetase activity. Comparative proteome analysis of a lysX mutant of Mycobacterium avium strain 104 and the wild type indicated that the lysX mutant strain undergoes a transition in phenotype by switching the carbon metabolism to ß-oxidation of fatty acids, along with accumulation of lipid inclusions. Surprisingly, proteins associated with intracellular survival were upregulated in the lysX mutant, even during extracellular growth, preparing bacteria for the conditions occurring inside host cells. In line with this, the lysX mutant exhibited enhanced intracellular growth in human-blood-derived monocytes. Thus, our study exposes the significance of lysX in the metabolism and virulence of the environmental pathogen M. avium hominissuis.


Subject(s)
Gene Expression Regulation, Bacterial , Lysine-tRNA Ligase/analysis , Metabolism , Mycobacterium avium/growth & development , Mycobacterium avium/metabolism , Proteome/analysis , Carbon/metabolism , Humans , Lipid Metabolism , Lysine-tRNA Ligase/deficiency , Monocytes/microbiology , Mycobacterium avium/chemistry , Mycobacterium avium/genetics , Oxidation-Reduction , Virulence
11.
J Immunol ; 199(4): 1429-1439, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28687660

ABSTRACT

IFN-γ is known to be predominantly produced by lymphoid cells such as certain subsets of T cells, NK cells, and other group 1 innate lymphoid cells. In this study, we used IFN-γ reporter mouse models to search for additional cells capable of secreting this cytokine. We identified a novel and rare population of nonconventional IFN-γ-producing cells of hematopoietic origin that were characterized by the expression of Thy1.2 and the lack of lymphoid, myeloid, and NK lineage markers. The expression of IFN-γ by this population was higher in the liver and lower in the spleen. Furthermore, these cells were present in mice lacking both the Rag2 and the common γ-chain (γc) genes (Rag2-/-γc-/-), indicating their innate nature and their γc cytokine independence. Rag2-/-γc-/- mice are as resistant to Mycobacterium avium as Rag2-/- mice, whereas Rag2-/- mice lacking IFN-γ are more susceptible than either Rag2-/- or Rag2-/-γc-/- These lineage-negative CD45+/Thy1.2+ cells are found within the mycobacterially induced granulomatous structure in the livers of infected Rag2-/-γc-/- animals and are adjacent to macrophages that expressed inducible NO synthase, suggesting a potential protective role for these IFN-γ-producing cells. Accordingly, Thy1.2-specific mAb administration to infected Rag2-/-γc-/- animals increased M. avium growth in the liver. Overall, our results demonstrate that a population of Thy1.2+ non-NK innate-like cells present in the liver expresses IFN-γ and can confer protection against M. avium infection in immunocompromised mice.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunity, Innate , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin Receptor Common gamma Subunit/immunology , Animals , Antibodies, Monoclonal/administration & dosage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Granuloma/immunology , Granuloma/microbiology , Immunocompromised Host/immunology , Interferon-gamma/immunology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Killer Cells, Natural/immunology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Liver/cytology , Liver/immunology , Liver/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mycobacterium avium/growth & development , Mycobacterium avium/immunology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Spleen/cytology , Spleen/immunology , Thy-1 Antigens/genetics , Thy-1 Antigens/immunology
13.
BMC Microbiol ; 16(1): 251, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27793104

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis infection is thought to induce oxidative stress. N-acetyl-cysteine (NAC) is widely used in patients with chronic pulmonary diseases including tuberculosis due to its mucolytic and anti-oxidant activities. Here, we tested whether NAC exerts a direct antibiotic activity against mycobacteria. METHODS: Oxidative stress status in plasma was compared between pulmonary TB (PTB) patients and those with latent M. tuberculosis infection (LTBI) or healthy uninfected individuals. Lipid peroxidation, DNA oxidation and cell death, as well as accumulation of reactive oxygen species (ROS) were measured in cultures of primary human monocyte-derived macrophages infected with M. tuberculosis and treated or not with NAC. M. tuberculosis, M. avium and M. bovis BCG cultures were also exposed to different doses of NAC with or without medium pH adjustment to control for acidity. The anti-mycobacterial effect of NAC was assessed in M. tuberculosis infected human THP-1 cells and bone marrow-derived macrophages from mice lacking a fully functional NADPH oxidase system. The capacity of NAC to control M. tuberculosis infection was further tested in vivo in a mouse (C57BL/6) model. RESULTS: PTB patients exhibited elevated levels of oxidation products and a reduction of anti-oxidants compared with LTBI cases or uninfected controls. NAC treatment in M. tuberculosis-infected human macrophages resulted in a decrease of oxidative stress and cell death evoked by mycobacteria. Importantly, we observed a dose-dependent reduction in metabolic activity and in vitro growth of NAC treated M. tuberculosis, M. avium and M. bovis BCG. Furthermore, anti-mycobacterial activity in infected macrophages was shown to be independent of the effects of NAC on the host NADPH oxidase system in vitro. Short-term NAC treatment of M. tuberculosis infected mice in vivo resulted in a significant reduction of mycobacterial loads in the lungs. CONCLUSIONS: NAC exhibits potent anti-mycobacterial effects and may limit M. tuberculosis infection and disease both through suppression of the host oxidative response and through direct antimicrobial activity.


Subject(s)
Acetylcysteine/pharmacology , Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Mycobacterium tuberculosis/drug effects , Adolescent , Adult , Animals , Case-Control Studies , Cell Death/drug effects , Cell Line , Disease Models, Animal , Humans , Latent Tuberculosis/blood , Latent Tuberculosis/drug therapy , Latent Tuberculosis/microbiology , Lipid Peroxidation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium avium/metabolism , Mycobacterium bovis/drug effects , Mycobacterium bovis/growth & development , Mycobacterium bovis/metabolism , NADPH Oxidases/deficiency , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Young Adult
14.
J Control Release ; 235: 112-124, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27261333

ABSTRACT

Tuberculosis (TB), a disease caused by the human pathogen Mycobacterium tuberculosis, recently joined HIV/AIDS on the top rank of deadliest infectious diseases. Low patient compliance due to the expensive, long-lasting and multi-drug standard therapies often results in treatment failure and emergence of multi-drug resistant strains. In this scope, antimicrobial peptides (AMPs) arise as promising candidates for TB treatment. Here we describe the ability of the exogenous AMP LLKKK18 to efficiently kill mycobacteria. The peptide's potential was boosted by loading into self-assembling Hyaluronic Acid (HA) nanogels. These provide increased stability, reduced cytotoxicity and degradability, while potentiating peptide targeting to main sites of infection. The nanogels were effectively internalized by macrophages and the peptide presence and co-localization with mycobacteria within host cells was confirmed. This resulted in a significant reduction of the mycobacterial load in macrophages infected in vitro with the opportunistic M. avium or the pathogenic M. tuberculosis, an effect accompanied by lowered pro-inflammatory cytokine levels (IL-6 and TNF-α). Remarkably, intra-tracheal administration of peptide-loaded nanogels significantly reduced infection levels in mice infected with M. avium or M. tuberculosis, after just 5 or 10 every other day administrations. Considering the reported low probability of resistance acquisition, these findings suggest a great potential of LLKKK18-loaded nanogels for TB therapeutics.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Antitubercular Agents/administration & dosage , Drug Carriers/administration & dosage , Gels/administration & dosage , Hyaluronic Acid/administration & dosage , Nanostructures/administration & dosage , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/therapeutic use , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Cell Survival/drug effects , Cells, Cultured , Drug Carriers/chemistry , Drug Carriers/therapeutic use , Gels/chemistry , Gels/therapeutic use , Hyaluronic Acid/chemistry , Hyaluronic Acid/therapeutic use , Interleukin-6/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Nanostructures/chemistry , Nanostructures/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Tumor Necrosis Factor-alpha/immunology
15.
Antimicrob Agents Chemother ; 60(4): 2577-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883697

ABSTRACT

Thein vitroactivities of clarithromycin and tigecycline alone and in combination againstMycobacterium aviumwere assessed. The activity of clarithromycin was time dependent, highly variable, and often resulted in clarithromycin resistance. Tigecycline showed concentration-dependent activity, and mycobacterial killing could only be achieved at high concentrations. Tigecycline enhanced clarithromycin activity againstM. aviumand prevented clarithromycin resistance. Whether there is clinical usefulness of tigecycline in the treatment ofM. aviuminfections needs further study.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clarithromycin/pharmacology , Drug Resistance, Bacterial , Minocycline/analogs & derivatives , Mycobacterium avium/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Microbial Sensitivity Tests , Minocycline/pharmacology , Mycobacterium avium/growth & development , Tigecycline , Time Factors
16.
Antimicrob Agents Chemother ; 60(2): 1097-105, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643335

ABSTRACT

Multidrug therapy is a standard practice when treating infections by nontuberculous mycobacteria (NTM), but few treatment options exist. We conducted this study to define the drug-drug interaction between clofazimine and both amikacin and clarithromycin and its contribution to NTM treatment. Mycobacterium abscessus and Mycobacterium avium type strains were used. Time-kill assays for clofazimine alone and combined with amikacin or clarithromycin were performed at concentrations of 0.25× to 2× MIC. Pharmacodynamic interactions were assessed by response surface model of Bliss independence (RSBI) and isobolographic analysis of Loewe additivity (ISLA), calculating the percentage of statistically significant Bliss interactions and interaction indices (I), respectively. Monte Carlo simulations with predicted human lung concentrations were used to calculate target attainment rates for combination and monotherapy regimens. Clofazimine alone was bacteriostatic for both NTM. Clofazimine-amikacin was synergistic against M. abscessus (I = 0.41; 95% confidence interval [CI], 0.29 to 0.55) and M. avium (I = 0.027; 95% CI, 0.007 to 0.048). Based on RSBI analysis, synergistic interactions of 28.4 to 29.0% and 23.2 to 56.7% were observed at 1× to 2× MIC and 0.25× to 2× MIC for M. abscessus and M. avium, respectively. Clofazimine-clarithromycin was also synergistic against M. abscessus (I = 0.53; 95% CI, 0.35 to 0.72) and M. avium (I = 0.16; 95% CI, 0.04 to 0.35), RSBI analysis showed 23.5% and 23.3 to 53.3% at 2× MIC and 0.25× to 0.5× MIC for M. abscessus and M. avium, respectively. Clofazimine prevented the regrowth observed with amikacin or clarithromycin alone. Target attainment rates of combination regimens were >60% higher than those of monotherapy regimens for M. abscessus and M. avium. The combination of clofazimine with amikacin or clarithromycin was synergistic in vitro. This suggests a potential role for clofazimine in treatment regimens that warrants further evaluation.


Subject(s)
Amikacin/pharmacology , Anti-Bacterial Agents/pharmacology , Clarithromycin/pharmacology , Clofazimine/pharmacology , Mycobacterium avium/drug effects , Nontuberculous Mycobacteria/drug effects , Drug Interactions , Drug Synergism , Drug Therapy, Combination , Microbial Sensitivity Tests , Monte Carlo Method , Mutation , Mycobacterium avium/genetics , Mycobacterium avium/growth & development , Nontuberculous Mycobacteria/genetics , Nontuberculous Mycobacteria/growth & development
17.
Nanotoxicology ; 10(6): 761-9, 2016 08.
Article in English | MEDLINE | ID: mdl-26618564

ABSTRACT

Antibiotic resistance is a serious problem. Nanotechnology offers enormous potential in medicine, yet there is limited knowledge regarding the toxicity of nanoparticles (NP) for mycobacterial species that cause serious human diseases (e.g. tuberculosis (TB) and leprosy). Mycobacterial diseases are a major global health problem; TB caused by Mycobacterium tuberculosis (Mtb) kills up to 2 million people annually and there are over 200 000 leprosy cases each year caused by Mycobacterium leprae (M. leprae). Few drugs are effective against these mycobacteria and increasing antibiotic resistance exacerbates the problem. As such, alternative therapies are urgently needed but most current assays used to assess the effectiveness of therapeutics against mycobacteria are slow and expensive. This study aimed to develop a rapid, low-cost assay which can be used for screening the antimicrobial properties of compounds against pathogenic mycobacteria and to assess the toxicity of three NP (silver [Ag], copper oxide [Cu(II)O], and zinc oxide [ZnO]) against a green fluorescent protein reporter strain of Mycobacterium avium subspecies paratuberculosis, a slow growing, pathogenic mycobacterial species causing paratuberculosis in ruminants. Fluorescence was used to monitor mycobacterial growth over time, with NP concentrations of 6.25-100 µg/mL tested for up to 7 days, and a method of data analysis was designed to permit comparison between results. Mycobacterial sensitivity to the NP was found to be NP composition specific and toxicity could be ranked in the following order: Ag > Cu(II)O > ZnO.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical/methods , Mycobacterium avium/drug effects , Nanoparticles/chemistry , Animals , Anti-Bacterial Agents/chemistry , Drug Resistance, Bacterial/drug effects , Green Fluorescent Proteins/genetics , Humans , Microbial Viability/drug effects , Mycobacterium avium/genetics , Mycobacterium avium/growth & development , Particle Size , Surface Properties
18.
Article in English | MEDLINE | ID: mdl-26380226

ABSTRACT

Mycobacterium avium subsp. hominissuis (MAH) is an environmental bacteria that infects immunocompromised humans. MAH cases are increasing in incidence, making it crucial to gain knowledge of the pathogenic mechanisms associated with the bacterium. MAH infects macrophages and after several days the infection triggers the phagocyte apoptosis. Many of the intracellular MAH escape the cell undergoing apoptosis leading to infection of neighboring macrophages. We screened a transposon bank of MAH mutants in U937 mononuclear phagocytes for the inability to escape macrophages undergoing apoptosis. Mutations in genes; MAV_2235, MAV_2120, MAV_2410, and MAV_4563 resulted in the inability of the bacteria to exit macrophages upon apoptosis. Complementation of the mutations corrected the phenotype either completely or partially. Testing for the ability of the mutants to survive in macrophages compared to the wild-type bacterium revealed that the mutant clones were not attenuated up to 4 days of infection. Testing in vivo, however, demonstrated that all the MAH clones were attenuated compared with the wild-type MAC 104 in tissues of mice. Although the mechanism associated with the bacterial inability to leave apoptotic macrophages is unknown, the identification of macrophage cytoplasm targets for the MAH proteins suggest that they interfere either with protein degradation machinery or post-translation mechanisms. The identification of tatC as a MAH protein involved in the ability of MAH to leave macrophages, suggests that secreted effector(s) are involved in the process. The study reveals a pathway of escape from macrophages, not shared with Mycobacterium tuberculosis.


Subject(s)
Apoptosis , Genes, Bacterial , Host-Pathogen Interactions , Macrophages/immunology , Macrophages/microbiology , Mycobacterium avium/immunology , Mycobacterium avium/physiology , Animals , Cell Line , DNA Transposable Elements , Disease Models, Animal , Gene Knockout Techniques , Genetic Complementation Test , Humans , Mice , Mutagenesis, Insertional , Mycobacterium avium/genetics , Mycobacterium avium/growth & development , Tuberculosis/microbiology , Tuberculosis/pathology , Virulence
19.
Proc Natl Acad Sci U S A ; 112(31): E4272-80, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195781

ABSTRACT

Several mechanisms are involved in controlling intracellular survival of pathogenic mycobacteria in host macrophages, but how these mechanisms are regulated remains poorly understood. We report a role for Kelch-like ECH-associated protein 1 (Keap1), an oxidative stress sensor, in regulating inflammation induced by infection with Mycobacterium avium in human primary macrophages. By using confocal microscopy, we found that Keap1 associated with mycobacterial phagosomes in a time-dependent manner, whereas siRNA-mediated knockdown of Keap1 increased M. avium-induced expression of inflammatory cytokines and type I interferons (IFNs). We show evidence of a mechanism whereby Keap1, as part of an E3 ubiquitin ligase complex with Cul3 and Rbx1, facilitates ubiquitination and degradation of IκB kinase (IKK)-ß thus terminating IKK activity. Keap1 knockdown led to increased nuclear translocation of transcription factors NF-κB, IFN regulatory factor (IRF) 1, and IRF5 driving the expression of inflammatory cytokines and IFN-ß. Furthermore, knockdown of other members of the Cul3 ubiquitin ligase complex also led to increased cytokine expression, further implicating this ligase complex in the regulation of the IKK family. Finally, increased inflammatory responses in Keap1-silenced cells contributed to decreased intracellular growth of M. avium in primary human macrophages that was reconstituted with inhibitors of IKKß or TANK-binding kinase 1 (TBK1). Taken together, we propose that Keap1 acts as a negative regulator for the control of inflammatory signaling in M. avium-infected human primary macrophages. Although this might be important to avoid sustained or overwhelming inflammation, our data suggest that a negative consequence could be facilitated growth of pathogens like M. avium inside macrophages.


Subject(s)
Inflammation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium avium/physiology , Signal Transduction , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cytokines/biosynthesis , Gene Knockdown Techniques , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/metabolism , Kelch-Like ECH-Associated Protein 1 , Mycobacterium avium/growth & development , NF-kappa B/metabolism , Phagosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Transport , Proteolysis , Reactive Oxygen Species/metabolism , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/pathology , Ubiquitination , Up-Regulation
20.
Nat Commun ; 6: 6211, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25648615

ABSTRACT

Most macrophages remain uninfected in HIV-1-infected patients. Nevertheless, the phagocytic capacity of phagocytes from these patients is impaired, favouring the multiplication of opportunistic pathogens. The basis for this phagocytic defect is not known. HIV-1 Tat protein is efficiently secreted by infected cells. Secreted Tat can enter uninfected cells and reach their cytosol. Here we found that extracellular Tat, at the subnanomolar concentration present in the sera of HIV-1-infected patients, inhibits the phagocytosis of Mycobacterium avium or opsonized Toxoplasma gondii by human primary macrophages. This inhibition results from a defect in mannose- and Fcγ-receptor-mediated phagocytosis, respectively. Inhibition relies on the interaction of Tat with phosphatidylinositol (4,5)bisphosphate that interferes with the recruitment of Cdc42 to the phagocytic cup, thereby preventing Cdc42 activation and pseudopod elongation. Tat also inhibits FcγR-mediated phagocytosis in neutrophils and monocytes. This study provides a molecular basis for the phagocytic defects observed in uninfected phagocytes following HIV-1 infection.


Subject(s)
HIV-1/physiology , Macrophages/drug effects , Monocytes/drug effects , Neutrophils/drug effects , Phagocytosis/drug effects , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Bystander Effect , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Mannose Receptor , Mannose-Binding Lectins/antagonists & inhibitors , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Monocytes/metabolism , Monocytes/ultrastructure , Mycobacterium avium/growth & development , Neutrophils/metabolism , Neutrophils/ultrastructure , Phosphatidylinositol 4,5-Diphosphate/antagonists & inhibitors , Phosphatidylinositol 4,5-Diphosphate/metabolism , Primary Cell Culture , Protein Transport/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/genetics , Receptors, IgG/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction , Toxoplasma/growth & development , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , tat Gene Products, Human Immunodeficiency Virus/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...